Contemporary Endocrinology Series Editor: Leonid Poretsky Anthony C. Hackney Naama W. Constantini Editors Endocrinology of Physical Activity and Sport Third Edition Contemporary Endocrinology Series Editor Leonid Poretsky Division of Endocrinology Lenox Hill Hospital New York, NY, USA More information about this series at http://www.springer.com/series/7680 Anthony C. Hackney Naama W. Constantini Editors Endocrinology of Physical Activity and Sport Third Edition Editors Anthony C. Hackney Department of Exercise & Sport Science, Department of Nutrition University of North Carolina Chapel Hill, NC USA Naama W. Constantini Heidi Rothberg Sport Medicine Center Shaare Zedek Medical Center Jerusalem Jerusalem Israel ISSN 2523-3785 ISSN 2523-3793 (electronic) Contemporary Endocrinology ISBN 978-3-030-33375-1 ISBN 978-3-030-33376-8 (eBook) https://doi.org/10.1007/978-3-030-33376-8 © Springer Nature Switzerland AG 2020 This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed. The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general use. The publisher, the authors and the editors are safe to assume that the advice and information in this book are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty, expressed or implied, with respect to the material contained herein or for any errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. This Humana imprint is published by the registered company Springer Nature Switzerland AG The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland This book is dedicated to my mentor, the late Professor Atko Viru, and my family who have always provided unwavering support for me. My since thanks and loving gratitude to each of you. – Anthony C. Hackney I dedicate this book to my late parents, Prof. RJZ Werblowsky and his beloved wife, Aliza, who passed away since the last edition. I owe them both my family and career. – Naama W. Constantini While finalizing this edition, Dr. Barbara Drinkwater, the pioneer of female athlete sports medicine and science research, has passed away. She will always be remembered as the first woman president of the American College of Sports Medicine and a leader to so many of us, especially in the field of eating disorders, menstrual irregularities, and bone health. The chapters in this book dealing with female physiology and endocrinology are honorably dedicated to her. – Naama W. Constantini and Anthony C. Hackney v Series Editor Foreword With the twin epidemics of obesity and diabetes upon us, endocrinologists often counsel patients on the benefits of exercise. During our many years of training, however, relatively little time is spent learning about the relationship between exercise and the endocrine system. To address this deficit, the volume edited by Drs. Anthony C. Hackney and Naama W. Constantini and written by an illustrious international group of experts provides an immense wealth of information on the topic. The relationship between exercise and the endocrine system is complicated and bidirectional: the effectiveness of exercise training in part depends on the state of the endocrine system, while all components of the endocrine system can be dramatically affected by exercise. The interactions between hormones and exercise extend well beyond the obvious (exercise and energy metabolism, exercise and diabetes) to include reproductive, adrenal and growth hormone axes, as well as bone metabolism, thyroid function, endogenous opiates, and circadian endocrine physiology, among others. Moreover, the relationship between the endocrine system and exercise evolves during the person’s lifetime (from childhood to puberty to advanced age) and is affected by both type and intensity of exercise (e.g., acute vs. chronic, moderate exercise vs. Olympic athlete training, etc.). Given these complexities, it is remarkable how clearly and completely the authors of this encyclopedic text have been able to cover their subject. The book is a true pleasure to read. It is exceptionally well referenced and, without a doubt, will become a valuable resource for anybody interested in human physiology. This most certainly includes endocrinologists, who, after becoming familiar with the content of this volume, will find themselves on a much firmer ground while advising their patients on the myriads of benefits, as well as some potential risks, of exercise. New York, NY, USA Leonid Poretsky, MD vii Preface The first edition of this book was entitled Sports Endocrinology edited by Michelle P. Warren and Naama W. Constantini and published in 2000. It was the first book with incursions into this complex and critically important topic area of exercise, sports, and hormones. It answered a recognized need and was well received by the scientific community. Twelve years later, the book took on a new expanded title, new editorship, and new authorship of chapter topics, and the second edition was published. It too was highly popular and a leading volume on the discipline. Now, after five additional years, a third edition has been developed with revised and updated content as well as new expanded materials. Nevertheless, over its evolution and multiple editions, the emphasis of the book has remained the same: to provide the reader with current, insightful discussion of the key elements of endocrinology as they relate to physical activity, exercise, and sports. Endocrinology is a demanding scientific endeavor and when overlaid with the unique aspects of the physical stress of exercise and exercise training can become a daunting topic. The editors are profoundly grateful to the contributor’s authors who have painstakingly and carefully crafted each of their discussions to aid the reader in overcoming what some might consider an insurmountable set of topics. The author’s scholarship, devotion to the scientific method, and overall professionalism have allowed for a new edition that not only reflects the present state of knowledge on each of their topics but will undoubtedly serve as a stimulus for further advances in this highly dynamic, constantly evolving, and challenging subject. Our sincere thanks to each of them for their efforts. We hope the readers will enjoy this new edition and it spurs them to ask new research questions. Chapel Hill, NC, USA Jerusalem, Israel Anthony C. Hackney, PhD, DSc Naama W. Constantini, MD ix Contents 1Methodological Considerations in Exercise Endocrinology�������� 1 Anthony C. Hackney, Abbie E. Smith-Ryan, and Julius E. Fink 2Endogenous Opiates and Exercise-­Related Hypoalgesia ������������ 19 Allan H. Goldfarb, Robert R. Kraemer, and Brandon A. Baiamonte 3The Effect of Exercise on the Hypothalamic-PituitaryAdrenal Axis ������������������������������������������������������������������������������������ 41 David H. St-Pierre and Denis Richard 4Impact of Chronic Training on Pituitary Hormone Secretion in Humans������������������������������������������������������������������������ 55 Johannes D. Veldhuis and Kohji Yoshida 5Exercise and the GH-IGF-I Axis���������������������������������������������������� 71 Alon Eliakim and Dan Nemet 6Exercise and Thyroid Function������������������������������������������������������ 85 Dorina Ylli, Joanna Klubo-Gwiezdzinska, and Leonard Wartofsky 7The Male Reproductive System, Exercise, and Training: Endocrine Adaptations�������������������������������������������� 109 Fabio Lanfranco and Marco Alessandro Minetto 8Exercise and the Hypothalamus: Ovulatory Adaptations������������ 123 Angela Y. Liu, Moira A. Petit, and Jerilynn C. Prior 9Adrenergic Regulation of Energy Metabolism������������������������������ 153 Michael Kjær and Kai Lange 10Sex Differences in Energy Balance and Weight Control�������������� 161 Kristin S. Ondrak 11Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function���������������� 171 Anne B. Loucks 12Ghrelin Responses to Acute Exercise and Training���������������������� 193 Jaak Jürimäe xi xii 13Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise���������������������������������������������������������� 209 Charles E. Wade 14Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone ������������������������������������������������������������ 229 Whitney R.D. Duff and Philip D. Chilibeck 15Interrelations Between Acute and Chronic Exercise Stress and the Immune and Endocrine Systems �������������������������� 249 Jonathan Peake 16Effects of Female Reproductive Hormones on Sports Performance�������������������������������������������������������������������� 267 Constance M. Lebrun, Sarah M. Joyce, and Naama W. Constantini 17Endocrine Implications of Relative Energy Deficiency in Sport �������������������������������������������������������������������������� 303 Katherine M. Cooper and Kathryn E. Ackerman 18Vitamin D and Exercise Performance�������������������������������������������� 321 Joi J. Thomas and D. Enette Larson-Meyer 19The Effects of Altitude on the Hormonal Response to Physical Exercise�������������������������������������������������������������������������� 341 Nunzia Prencipe, Chiara Bona, Fabio Lanfranco, Silvia Grottoli, and Andrea Silvio Benso 20An Introduction to Circadian Endocrine Physiology: Implications for Exercise and Sports Performance���������������������� 363 Teodor T. Postolache, Arshpreet Gulati, Olaoluwa O. Okusaga, and John W. Stiller 21The Role of Hormones in Exercise-­Induced Muscle Hypertrophy������������������������������������������������������������������������ 391 Julius E. Fink 22Endocrine Responses to Acute and Chronic Exercise in the Developing Child ������������������������������������������������������������������ 399 Daniela A. Rubin 23Exercise in Older Adults: The Effect of Age on Exercise Endocrinology���������������������������������������������������������������������������������� 421 Jennifer L. Copeland 24Immune, Endocrine, and Soluble Factor Interactions During Aerobic Exercise in Cancer Survivors������������������������������ 441 Elizabeth S. Evans, Erik D. Hanson, and Claudio L. Battaglini 25Type I Diabetes and Exercise���������������������������������������������������������� 459 Sam N. Scott, Michael C. Riddell, and Jane E. Yardley Contents Contents xiii 26Extreme Sports and Type 1 Diabetes Mellitus in the Twenty-­First Century: The Promise of Technology���������������������� 483 Karen M. Tordjman and Anthony C. Hackney 27The Endocrine System in Overtraining ���������������������������������������� 495 David R. Hooper, Ann C. Snyder, and Anthony C. Hackney 28Hormones as Performance-­Enhancing Agents������������������������������ 507 Erick J. Richmond and Alan D. Rogol 29Metabolic Syndrome, Hormones, and Exercise���������������������������� 519 Konstantina Dipla, Andreas Zafeiridis, and Karen M. Tordjman 30Exercise and Training Effects on Appetite-Regulating Hormones in Individuals with Obesity������������������������������������������ 535 Hassane Zouhal, Ayoub Saeidi, Sarkawt Kolahdouzi, Sajad Ahmadizad, Anthony C. Hackney, and Abderraouf Ben Abderrahmane Index���������������������������������������������������������������������������������������������������������� 563 Contributors Abderraouf Ben Abderrahmane, PhD Higher Institute of Sport Sciences and Physical Education of Ksar Saïd, Department of Biological Sciences, Ariana, Tunisia Kathryn E. Ackerman, MD, MPH Harvard Medical School, Boston Children’s Hospital, Department of Sports Medicine and Endocrinology, Boston, MA, USA Sajad Ahmadizad, PhD Department of Biological Sciences in Sport and Health, Faculty of Sports Sciences and Health, Shahid Beheshti University, Tehran, Iran Brandon A. Baiamonte, PhD Southeastern Department of Psychology, Hammond, LA, USA Louisiana University, Claudio L. Battaglini, PhD Department of Exercise & Sport Science, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA Andrea Silvio Benso, MD, PhD AOU Citta della Salute e della Scienza di Torino, Division of Endocrinology, Diabetology and Metabolism, University of Turin, Department of Medical Sciences, Turin, Italy Chiara Bona, MD AOU Citta della Salute e della Scienza di Torino, Division of Endocrinology, Diabetology and Metabolism, University of Turin, Department of Medical Sciences, Turin, Italy Philip D. Chilibeck, PhD University of Saskatchewan, College of Kinesiology, Saskatoon, SK, Canada Naama W. Constantini, MD, DFM Heidi Rothberg Sport Medicine Center, Department of Sport Medicine, Shaare Zedek Medical Center Jerusalem, affiliated with the Hebrew University School of Medicine, Jerusalem, Israel Katherine M. Cooper, BA University of Massachusetts Medical School, Worcester, MA, USA Jennifer L. Copeland, PhD Department of Kinesiology, University of Lethbridge, Lethbridge, AB, Canada Konstantina Dipla, PhD Department of Sport Science, TEFAA SERRON, Aristotle University of Thessaloniki, Serres, Greece xv xvi Patricia Katherine Doyle-Baker, DrPH, MA, BSc University of Calgary, Human Performance Lab, Faculty of Kinesiology, Calgary, AB, Canada Whitney R.D. Duff, PhD University of Saskatchewan, College of Kinesiology, Saskatoon, SK, Canada Alon Eliakim, MD Pediatric Department and Endocrinology Clinic, Meir Medical Center, Sackler School of Medicine, Tel Aviv University, Department of Pediatrics, Kfar Saba, Israel Elizabeth S. Evans, PhD Elon University, Physical Therapy Education, Elon, NC, USA Julius E. Fink, PhD Juntendo University Graduate School of Medicine, Department of Urology, Tokyo, Japan Allan H. Goldfarb, PhD University of North Carolina Greensboro, Department of Kinesiology, Greensboro, NC, USA Silvia Grottoli, MD AOU Citta della Salute e della Scienza di Torino, Division of Endocrinology, Diabetology and Metabolism, University of Turin, Department of Medical Sciences, Turin, Italy Arshpreet Gulati, MD University of Maryland Medical Centre, Mood and Anxiety Program, Baltimore, MD, USA St. Elizabeths Hospital, Department of Neurology Consultation Service, Washington, DC, USA Anthony C. Hackney, PhD, DSc Department of Exercise & Sport Science, Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA Erik D. Hanson, PhD Department of Exercise & Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA David R. Hooper, PhD Jacksonville University, Department of Kinesiology, Jacksonville, FL, USA Sarah M. Joyce, BExSc Griffith Health Institute, Gold Coast, QLD, Australia Jaak Jürimäe, PhD Institute of Sport Sciences and Physiotherapy, University of Tartu, Tartu, Estonia Michael Kjær, MD, PhD Department of Clinical Medicine, BispebjergFrederiksberg Hospital, Copenhagen, Denmark Joanna Klubo-Gwiezdzinska, MD, PhD, MHSc National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Disease/ Metabolic Disease Branch, Bethesda, MD, USA Sarkawt Kolahdouzi, PhD Exercise Biochemistry Division, Department of Exercise Physiology, Faculty of Sport Sciences, University of Mazandaran, Babolsar, Mazandaran, Iran Contributors Contributors xvii Robert R. Kraemer, EdD Southeastern Louisiana University, Kinesiology and Health Studies, Hammond, LA, USA Fabio Lanfranco, MD, PhD AOU Citta della Salute e della Scienza di Torino, Division of Endocrinology, Diabetology and Metabolism, University of Turin, Department of Medical Sciences, Turin, Italy Kai Lange, MD Department of Clinical Medicine, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark D. Enette Larson-Meyer, PhD Department of Family and Consumer Services, University of Wyoming, Laramie, WY, USA Constance M. Lebrun, MDCM, MPE Department of Family Medicine, Kaye Edmonton Clinic, Glen Sather Sports Medicine Clinic, University of Alberta, Edmonton, AB, Canada Angela Y. Liu, MD University of British Columbia, Department of Endocrinology, Vancouver, BC, Canada Anne B. Loucks, PhD Biological Sciences, Ohio Musculoskeletal and Neurological Institute, Ohio University, Athens, OH, USA Marco Alessandro Minetto, MD, PhD Division of Physical Medicine and Rehabilitation, Department of Surgical Sciences, University of Turin, Turin, Italy Dan Nemet, MD Child Health and Sports Center, Meir Medical Center, Sackler School of Medicine, Tel Aviv University, Department of Pediatrics, Kfar Saba, Israel Olaoluwa O. Okusaga, MD Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, Houston, TX, USA Kristin S. Ondrak, PhD Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC, USA Jonathan Peake, PhD School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia Moira A. Petit, PhD Activ8, LLC, St. Paul, MN, USA Teodor T. Postolache, MD University of Maryland Medical Centre, Mood and Anxiety Program, Baltimore, MD, USA The Center for Sleep, Mood, Anxiety, and Performance, Washington, DC, USA Nunzia Prencipe, MD AOU Citta della Salute e della Scienza di Torino, Division of Endocrinology, Diabetology and Metabolism, University of Turin, Department of Medical Sciences, Turin, Italy Jerilynn C. Prior, BA, MD University of British Columbia, Medicine, Division of Endocrinology and Metabolism, Vancouver, BC, Canada xviii Denis Richard, PhD IUCPQ Research Centre, Laval University, Quebec City, QC, Canada Erick J. Richmond, MD, MSc National Children’s Hospital, Pediatric Endocrinology, San Jose, Costa Rica Michael C. Riddell, PhD York University, School of Kinesiology and Health Sciences, Toronto, ON, Canada Alan D. Rogol, MD, PhD University of Virginia Medical Center, Department of Pediatrics, Charlottesville, VA, USA Daniela A. Rubin, PhD Department of Kinesiology, California State University Fullerton, Fullerton, CA, USA Ayoub Saeidi, PhD Department of Biological Sciences in Sport and Health, Faculty of Sports Sciences and Health, Shahid Beheshti University, Tehran, Iran Sam N. Scott, PhD York University, School of Kinesiology and Health Sciences, Toronto, ON, Canada Abbie E. Smith-Ryan, PhD Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC, USA Ann C. Snyder, PhD University of Wisconsin – Milwaukee, Department of Kinesiology, Milwaukee, WI, USA John W. Stiller, MD Neurology Consultation Service, St. Elizabeths Hospital/DC Department of Behavioral Health, Department of Neurology, Washington, DC, USA David H. St-Pierre, PhD University of Quebec at Montreal (UQAM), Montreal, QC, Canada Joi J. Thomas, MS Department of Athletics, University of Wyoming, Laramie, WY, USA Karen M. Tordjman, MD Tel Aviv Sourasky Medical Center, Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Institute of Endocrinology, Metabolism, and Hypertension, Tel Aviv, Israel Johannes D. Veldhuis, MD Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA Charles E. Wade, PhD Center for Translational Injury Research (CeTIR), Houston, TX, USA Leonard Wartofsky, MD Thyroid Cancer Research, Georgetown University School of Medicine, MedStar Health Research Institute, Department of Endocrinology, Washington, DC, USA Jane E. Yardley, PhD University of Alberta, Augustana Faculty, Camrose, AB, Canada Contributors Contributors xix Dorina Ylli, MD, PhD MedStar Health Research Institute, Thyroid Cancer Research Center, Washington, DC, USA Kohji Yoshida, MD Department of Obstetrics and Gynecology, University of Occupational and Environmental Health, Kitakyushu, Japan Andreas Zafeiridis, PhD Department of Sport Science, TEFAA SERRON, Aristotle University of Thessaloniki, Serres, Greece Hassane Zouhal, PhD Univ Rennes, M2S (Laboratoire Mouvement, Sport, Santé), Rennes, France 1 Methodological Considerations in Exercise Endocrinology Anthony C. Hackney, Abbie E. Smith-Ryan, and Julius E. Fink Introduction Over the last several decades, an increasing number of exercise science investigations have incorporated measurements of endocrine function (e.g., hormones, cytokines) into their research designs and protocols [1, 2]. This approach has allowed for a heightened level of investigation into research which examines the physiological mechanisms associated with clinical and performance-­related conditions found in individuals involved in exercise training. Some exercise science investigations, however, have not always controlled certain critical factors (e.g., time of day for blood sampling, level of chronic training, etc.) that can influence many of the hormones associated with the human endocrine system. This lack of investigative control has often resulted in the resulting research findings to be inconsistent, contradictory, and A. C. Hackney (*) Department of Exercise & Sport Science, Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA e-mail: ach@email.unc.edu A. E. Smith-Ryan Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC, USA J. E. Fink Juntendo University Graduate School of Medicine, Tokyo, Japan sometimes extremely difficult to interpret. This insufficient control of biological experimental factors appears to be due in part to limited knowledge by exercise science researchers in the area of clinical endocrine methodology and techniques. Experts suggest that the factors that influence hormonal measurements, and contribute to variance in experimental outcomes, can be categorized as consisting of two potential sources: factors affecting physiological variation (i.e., affiliated with the physiological function status of the subject) and factors affecting procedural-­ analytical variation (i.e., determined by the investigators conducting research) [1, 3]. Regardless of the source of variance, subject, or investigator derived, if it is not controlled or accounted for appropriately, the resulting hormonal measurements obtained can be compromised and thus call into question the scientific validity of a research study. The focus of this chapter is to provide background information for exercise science researchers on those physiological-procedural-analytical factors that can potentially affect endocrine measurements. The intent is for this material to serve as an introductory “fundamental coverage” on this topic in hopes of improving the quality of research in exercise endocrinology. The field of endocrinology uses numerous abbreviations for the many of the hormones that exist. To aid those researchers unfamiliar with © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_1 1 A. C. Hackney et al. 2 Table 1.1 The following are abbreviations commonly used for various hormones seen in exercise science and sport medicine endocrinological research (see Ref. [4]) Sex/Gender It appears that until the onset of puberty, there is little difference between males and females in their resting hormonal profile. Once puberty is reached though, there is increased androgenic steroid hormone production in the male, and the female starts the characteristic menstrual cycle pulsatile release of gonadotropin and sex steroid hormones [5–7]. Additionally, at puberty, resting leptin (an adipocyte cytokine; a low molecular weight protein that has endocrine-like actions on select physiological process such as the immune system [8]) levels tend to become increased in females, as compared to those in males [9]. In adulthood, the hormonal differences that begin to manifest at puberty tend to remain until females reach the postmenopausal period and males reach andropause [8, 9]. There are some sex-specific differences in the hormonal responses to exercise in males and females. These include an earlier and greater rise in testosterone and creatine kinase during exercise and up to 24 hours after exercise in males as compared to females [1, 10, 11] and a greater pre-exercise growth hormone response in females. Furthermore, the magnitude of the sex steroid hormonal response to exercise in females is influenced by the status and phase of their menstrual cycle [10, 12]. Interestingly, the menstrual cycle hormones can influence other hormones and their response to exercise (e.g., this lexicon, Table 1.1 lists those abbreviations increased estradiol-β-17 → increases growth horfor the most common hormones associated with mone levels) [10–13] (see later discussion conthe area of exercise science [4]. cerning the menstrual cycle in this chapter, as well as Chap. 16 in this book). On the other hand, some hormones show little or no differences in response to exercise between the sexes (e.g., Physiological Factors water balance hormones such as aldosterone and As mentioned, factors that can influence hor- arginine vasopressin) [5, 10, 12]. Due to these potential differences in outcomes monal measurements can be categorized into two broad areas: “physiological” and “procedural-­ due to sex, the researcher should be cautious analytical.” The physiological factors are those when using adult subject populations involving a that are determined to be connected in some way mixture of males and females in their studies. To to a biological function or status of the research avoid confounding results, researchers need to be subject (patient) at the time of the collection of the certain that the hormonal outcomes they are measpecimen (e.g., blood) to be analyzed. These are suring are not influenced by sex/gender and stage factors that can be viewed as pertaining to a vari- of menstrual cycle, as will be discussed in subsequent sections of this chapter. ety of endogenous aspects of the subject/patient. Name Adrenocorticotropic hormone Aldosterone Antidiuretic hormone Atrial natriuretic peptide Arginine vasopressin β-Endorphin Catecholamines Corticotropin-releasing hormone Cortisol Epinephrine Estradiol-β-17 Follicle-stimulating hormone Glucagon Gonadotropin-releasing hormone Growth hormone Growth hormone-releasing hormone Insulin Insulin-like growth factor [1] Leptin Luteinizing hormone Norepinephrine Parathyroid hormone Progesterone Prolactin Reverse triiodothyronine Testosterone Thyrotropin-releasing hormone Thyroid-stimulating hormone Thyroxine Triiodothyronine Abbreviations ACTH ALD ADH ANP AVP β-END Cats CRH CORT EPI E2 FSH GLU GnRH GH GHRH IN IGF1 LP LH NEPI PTH P PRL rT3 TEST TRH TSH T4 T3 1 Methodological Considerations in Exercise Endocrinology Age If subjects are not matched for age and maturity level, whenever possible, variance in the outcomes can be potentially increased. For example, a prepubertal and postpubertal child (of the same gender) will not typically display the exact same hormonal exercise responses or relationships [14, 15]. This is illustrated by the welldocumented increase in insulin resistance which is observed as an adolescent goes through puberty [16]. This concern should also be extended to the other end of the age spectrum. That is, a postmenopausal female or andropausal male could have drastically different hormonal responses when compared to a relative prepausal individual. For example, basal growth hormone and testosterone typically decrease with age, while cortisol and insulin resistance increase [17–19]. These types of age-related differences cannot only exist at rest, but also in response to exercise, and even after completing an exercise training program. As an illustration, a study by Brook et al. showed that the anabolic responses to resistance training are impaired within the elderly, possibly due to lower testosterone levels (young subjects = 367 ± 19; old subjects = 274 ± 19 ng•dl−1), ribosomal biogenesis (RNA:DNA ratio and c-MYC induction; young = +4 ± 2-fold change; old = +1.9 ± 1-fold change), and/or translational efficiency S6K1 phosphorylation (young = +10 ± 4-fold change; old = +4 ± 2-fold change) (see Chap. 21 in this book concerning S6K1 [mTOR substrate S6 kinase 1]) [20]. For this reason, it is important to match subjects in research studies by chronological age and/or maturation level in order to increase the homogeneity of the responses and decrease interindividual variability, obviously, that is, unless the researcher is trying to study agerelated changes among groups of individuals [3]. Ethnicity and Race A variety of different humoral constituents are known to vary between people of different races and ethnic groups [1, 3]. For example, resting 3 parathyroid hormone levels tend to be higher in Black compared to Caucasian individuals [21]. Caucasian females tend to have higher levels of estrogens than Asian females [1, 22]. Evidence also suggests that reproductive hormone levels during gestational periods may vary greatly across several races and ethnic groups (Caucasians, Blacks, Latinos, Asians, and Indians) [22–25]. Findings of greater resting insulin and degree of insulin resistance in certain Native American tribes (e.g., Pima Indians) have also been reported; however these differences may in fact be more related to obesity issues in these individuals [26]. Testosterone levels seem also to have differences among ethnicities, with Asian- and Indian-related ethnicities showing slightly lower levels as compared to other ethnicities [27]. Hormonal responses to exercise and exercise training related to race and ethnicity have not been well studied, and the limited available findings do not suggest drastically different response outcomes beyond basal differences. Further research is certainly necessary and warranted in this area [1, 25, 26]. Body Composition: Adiposity The level of adiposity of the body can greatly influence the release of certain cytokines by adipose tissue [3, 8, 9]. These cytokines in turn can have autocrine-, paracrine-, and endocrinelike actions and influence aspects of metabolism, reproductive, and inflammatory function [2, 3, 8, 9]. For example, increase in adipose tissue raises the expression of aromatase, triggering higher conversion rates of testosterone to estradiol, triggering a negative feedback to the pituitary gonadotropin secretion, and ultimately resulting in lower testosterone levels [28]. Additionally, several of these cytokines have been directly linked to the promotion of increased hormonal levels (e.g., increased interleukin-6 → increased cortisol) [8]. This situation becomes compounded as adiposity reaches the level of obesity and subsequently affects many hormones to a far greater degree. A. C. Hackney et al. 4 For example, insulin and leptin levels tend to be appreciably elevated at rest in many obese persons [29–33]. As levels of adiposity increase, the hormonal response to exercise and exercise training can change considerably from that of a normal-­ weight person. As an illustration, in obese persons, catecholamine and growth hormone response to exercise becomes blunted [33]. Cortisol responses to exercise seem to become elevated in some overweight-obese individuals, although isolated cases have shown cortisol responses have been shown to be blunted and reduced [32, 33]. Exercise training often allows a loss of body mass, in particular fat mass, which helps to normalize these hormones with levels observed in normal-weight people [33–37]. To ensure that varying levels of body composition of subjects will not confound hormonal outcomes, investigators need to match their subjects for adiposity as closely as possible and not just use body weight matching as criterion. Exactly how close of a match is needed is not known, but grouping normal-weight, overweight (body mass index (BMI) ≥ 25.0–< 30.0 kg/m2), and obese (BMI ≥ 30.0 kg/m2) individuals into the same subject group can most certainly complicate and add variance to some hormonal outcomes [1, 33]. Disease States Several disease conditions such as HIV, testicular cancer, hormonal disorders (e.g., Cushing’s syndrome, Graves’ disease, etc.), infections, chronic liver or kidney diseases, type 2 diabetes, and obesity have been shown to affect hormones; e.g., lower sex hormone levels [38, 39]. Specifically, infection-type diseases may lead to testicular dysfunction, and metabolic conditions may lead to hypogonadism via increased adipose tissue and inflammation [28]. Indeed, in HIVinfected patients, lymphoma, or syphilis effects on the pituitary, can trigger or mimic apoplexy and meningeal or pituitary infection leading to fibrosis and ultimately dysfunction [40]. Increased adipose tissue often observed in obese and patients with type 2 diabetes leads to increased aromatization activity converting testosterone to estradiol, leading to a negative feedback to the pituitary gonadotropin secretion triggering hypogonadism [28]. Many individuals are unaware of these conditions when signing up for a study; therefore thorough medical screening is an important consideration. Mental Health Select mental health conditions and states are associated with high levels of anxiety and apprehension (e.g., posttraumatic stress disorder), which can lead to enhanced activity of the sympathetic nervous system and hypothalamic-­pituitary-­ adrenal axis [41–43]. Subsequently, resting levels of circulating catecholamines, adrenocorticotropic hormone, β-endorphin, and cortisol can be elevated. In contrast, persons who are experiencing depression can have low arousal levels, and the abovementioned hormones could be suppressed. Moreover, depression is sometimes accompanied by low activity levels in the hypothalamic-pituitary-thyroid axis (i.e., low thyrotropin-releasing hormone, thyroid-­ stimulating hormone, thyroxine, and triiodothyronine) creating a euthyroid sick syndrome response [41–43]. These alterations in resting hormonal levels from such conditions can in turn result in altered hormonal responses to exercise and exercise training in individuals who have high levels of anxiety [44–46]. In some cases this can result in heightened responses (excessive) or diminished responses [44–46]. Evaluating the mental health status, via the completion of a screening questionnaire by a participant, can serve as an excellent tool to determine if a potential emotional or psychological problem exists which could confound hormonal measurements. A variety of such screening tools are available, and the reader is directed to several excellent references for overviews of this topic [47, 48]. Importantly, it is highly advisable that any such assessment be performed by a trained, qualified individual. 1 Methodological Considerations in Exercise Endocrinology Menstrual Cycle Menstrual status (eumenorrheic vs. oligomenorrheic vs. amenorrheic) and cycle phase (follicular, ovulation, luteal) in females can produce basal changes in key reproductive hormones such as estradiol-β-17, progesterone, luteinizing hormone (LH), testosterone, and follicle-stimulating hormone (FSH). These changes can be large and dramatic within select individuals. For example, the ovulatory and luteal phases result in increases in all of the aforementioned hormones above what is seen in the follicular phase (e.g., 2–10-fold greater in eumenorrheic female) [49]. These typical changes are depicted in Fig. 1.1. As noted earlier, select reproductive hormones (sex steroids) at rest can influence certain other nonreproductive hormones and nonreproductive physiological function such as estradiol-β-17 enhancing growth hormone release and thus subsequently increased lipid metabolism [11, 50, 51]. The menstrual status and cycle phase hormonal influences can carry over to have an impact on exercise and exercise training responses, too. Consequently, researchers may need to conduct exercise testing with females of similar menstrual status and/or in similar phases of their cycle. This precaution is also applicable to females who are using oral contraceptives, which can mimic some hormonal fluctuations similar to cycle phase changes [51, 52]. The precise impact of oral conceptive (OC) depends upon the composition of the OC used (mono-, bi-, or triphasic) and the dosage of the active estrogen and progestin agents in the pharmaceuticals. It is also an important consideration when there is an absence of a menstrual cycle either due to amenorrhea (especially hypothalamic based) or due to pregnancy, this absence leads to changes in hormonal levels and exercise responses [1, 2, 24]. Circadian Rhythms Over the course of a 24-h period, many hormonal levels will fluctuate and display circadian variations (see Chap. 20 of this book). In some cases these variances are due to pulse generator aspects, which is the spontaneous release of select hypothalamic hormonal releasing factors/hormones [53] within the endocrine regulatory axis. In other cases, variances are related to humoral stimuli, changes brought on by individual behavior or environmental factors, and these humoral stimuli influence hormonal release [54, 55]. Circadian hormones can display dramatic changes in levels due to their rhythm patterns, cortisol being a prime example. Morning cortisol levels are typically twice that of those found later Pituitary hormones = LH, FSH Ovarian hormones = Estrogen Progesterone Relative Hormonal Changes Fig. 1.1 Typical hormone changes (arbitrary scaling for concentration changes) associated with the menstrual cycle in eumenorrheic women 5 LH Progesterone Day 1 Menses (~ 3-5 days) Estrogen FSH Day 14 Ovulation Follicular Phase Day 28 Luteal Phase A. C. Hackney et al. 6 Table 1.2 Hormones that display discernable circadian patterns. The arrows indicate a relative direction for changes in concentration levels Hormone ACTH Aldosterone Cortisol AM concentration ↑ Early AM ↓ ↑↑ PM concentration ↓ ↑ ↓↓ Growth hormone LH-FSH ↑ Early AM ↓↑ ↓ ↓↑ Melatonin Parathyroid hormone Prolactin ↑ Early AM ↓ ↓ ↑ ↑ Early AM ↓ Late AM ↑ ↑ ↓ Testosterone Remarks Highest levels may be during sleep Highest levels may be during sleep Influenced by food intake; highest levels may be during sleep Only slight differences; highest levels may be during sleep Pulsatility or release and menstrual cycle phase override circadian pattern Highest levels may be during sleep Highest levels may be during sleep Highest levels may be during sleep Lessens with age Arrows indicate an increase (↑) or decrease (↓) in hormone concentration in the day [56–58]. Table 1.2 provides some reference on the circadian pattern seen in some key hormones. These fluctuations and circadian variations need to be addressed when conducting exercise research. Studies demonstrate that the magnitude of exercise responses may not be similar at different times of the day, even if the exercise intensity and duration are held constant [1, 56]. Investigators should plan accordingly so as to more carefully control and replicate the time of day in which research evaluations are conducted and hormonal specimen collected [59, 60]. otal Versus Free Hormone T Concentration A number of hormones exist in the circulation as either in there free or total amounts forms, the latter being the sum of the free and the carrierbound portion of the hormone. Steroid hormones are the principal example of this situation. Some investigators do not completely recognize this point and in conducting their research at times measure the wrong form of the hormone in question. A prime illustration of this is the hormone testosterone in which the free form is viewed as more biologically active. That is, the major part of circulating testosterone binds to sex hormone-binding globulin (SHBG) and some to albumin, leaving only a small fraction of testosterone as free form. In order to be bioactive, testosterone has to be unbound, making only free and albumin-bound (weak bound easy to dissociate) available for tissue uptake. It has been debated for a long time which form of testosterone (total vs. free) is a better indicator for testosterone levels in subjects. A recent study demonstrated that even if total testosterone levels are normal, low free testosterone is associated with hypogonadal signs and symptoms. This suggests free testosterone might be a more accurate measure of androgen-related conditions as compared to total testosterone, although clinicians/researchers must make their own determination on which hormonal form they should be accessing [61]. Procedural-Analytical Factors The second category of factors influencing hormonal measurements is made up of those factors that have procedural or analytical aspects to them. These factors are determined, selected, or in some way potentially controlled for by the investigators conducting or the participant involved with the research [1]. These factors can be viewed as exogenous relative to their influence. 1 Methodological Considerations in Exercise Endocrinology Ambient Environment When conducting research investigations, it is important to remember that excessive exposure to hot or cold ambient temperatures can stimulate the release of various hormones, e.g., those involved in water balance (aldosterone) or energy substrate mobilization (cortisol) [44, 62, 63]. Even elevated ambient relative humidity (water vapor) can induce this effect, primarily due to a compromised heat dissipation through reduced evaporative efficiency adding to the body core temperature [63]. These effects can be further augmented if hypoxemia is induced along with temperature extremes (e.g., mountain climbing), as can occur when moving to higher elevations and being exposed to greater degrees of hypoxia [64–66]. Many of the exercise and exercise training hormonal responses are tremendously impacted by environmental factors. In particular, catecholamines, growth hormone, aldosterone, arginine vasopressin, adrenocorticotropic hormone, and cortisol are all susceptible to changes in environmental conditions and show highly exacerbated responses in such varying conditions [1, 44, 62, 63]. To minimize these influences, it is critical to conduct exercise testing in controlled, standardized conditions such as in a laboratory. On the other hand, if conducting field research (where environmental standardization can be impossible), then it is important to measure/record environmental factors and convey them in any subsequent reporting of the data in the literature. Nutrition The nutritional status and practices of a research subject, including food composition, caloric intake, and timing of meals, can greatly impact the hormones associated with energy substrate mobilization and utilization (e.g., insulin, glucagon, epinephrine, growth hormone, insulin-like growth factor, cortisol) [1, 67, 68]. The exact nature of the effect (augmented or attenuation) depends on the interaction of the nutritional fac- 7 tors just mentioned and how severely the alterations are from the normal nutritional regimes of the individual [1, 33, 41]. The hormones noted above are critical during exercise to ensure that energy metabolism meets the demands of exercise. Thus, altered dietary practices and nutrition status of a subject can alter energy substrate (glycogen) storage and availability [68–70]. This in turn can cause the hormonal response to exercise to vary to some degree. For example, Galbo and associates demonstrated that the glucagon, epinephrine, growth hormone, and cortisol response to exercise were greater when a low-carbohydrate, high-fat diet is consumed (i.e., 4 days of consumption) compared to a normal mixed diet [67]. Normally in clinical settings, it is recommended that subjects be fasted prior to blood hormonal evaluations (e.g., 8 h). It is not always practical, however, for athletes to comply with such request due to their high demand for adequate caloric intake to maintain energy balance, anabolism, and muscle glycogen reserves. Therefore, a modified fasted approach may be necessary for this special population such as only a 4–6-h fast. Even with the constraints of working around an athlete’s special needs, it is still advisable that exercise investigators try to control and standardize the dietary practices of their subjects as much as possible to mitigate the effects of differing diet between subjects, and within an individual subject’s diet, if a repeated measures research design is being used [41, 67]. Nutrient Timing The concept of nutrient timing is arguably one of the most important aspects to account for when designing a study and evaluating results [71]. The evaluation of timing food consumption has been shown to influence muscle morphology outcomes directly and indirectly by stimulating hormone secretion [72]. As coined by Dr. John Ivy, the nutrient timing system accounts for three phases: the energy phase, anabolic phase, and growth phase [71]. Additional consideration should be given to the pre-exercise phase, which can largely influence the endocrine response during and 8 A. C. Hackney et al. can enhance muscle glycogen resynthesis and is enhanced with a protein/carbohydrate combination [72, 79]. A carbohydrate-amino acid supplement influenced testosterone and cortisol levels 120 min after intake and exercise [80]. However, post-exercise nutrient consumption consumed later following exercise (i.e., 8–9 h) has demonPre-exercise Cortisol levels, which help to strated no hormonal influence [81]. Intake of carmaintain the integrity of the immune system, are bohydrates + protein + vitamins post-exercise strongly influenced by glucose availability [73– has been shown to reduce free radicals and main75]. Additionally, acute carbohydrate intake can tain immune function. This may be a considerstimulate an increase in insulin and glucose lev- ation for researchers evaluating exercise and els, sparing muscle glycogen as well as reducing immunology characteristics, as well as various cortisol levels. Acute consumption of a glucose-­ aspect of overtraining. electrolyte solution (GES) prior to exercise has been shown to significantly reduce cortisol lev- Meal Frequency and Patterning els, when compared to water. Allowing a subject Meal frequency and overall caloric consumpto consume a carbohydrate drink before testing, tion may also influence metabolic-hormonal independent of amount (e.g., 25 g vs. 200 g), may markers, such as C-reactive protein, fasting significantly maintain glucose and cortisol levels plasma glucose, insulin, as well as total cholespost-exercise, as well as stabilizing the neutro- terol [82, 83]. In as much, investigators may phil to lymphocyte ratio [75]. Pre-exercise vita- consider questioning participants about food min consumption, or an antioxidant enhanced consumption patterns or utilize a food frequency beverage, may protect against acute tissue dam- questionnaire. age augmenting exercise adaptations and when consumed chronically may maintain immune Eating Disorders The eating disorder “anorexia nervosa” is a spesystem markers [76]. cial concern relative to nutrition status due to its Anabolic Phase (During Exercise) Carbohydrate profound effect on the endocrine system [1, 52, supplementation during exercise has also been 84]. Anorexics tend to have lower resting luteinassociated with a blunted cortisol, growth hor- izing hormone, follicle-stimulating hormone, and mone, and cytokine response while also main- estradiol-β-17 levels [84]. Anorexia also affects taining glucose levels and insulin stability [77, the pituitary-thyroid-glandular axis. Specifically, 78]. There is additional evidence demonstrating the condition is associated with suppression of reduced T cell and NK cell levels with carbohy- triiodothyronine, somewhat decreased thyroxine, drate feeding during exercise [77]. Acute, uncon- elevation of reverse triiodothyronine, and, occatrolled feedings should be accounted for when sionally, decreases in thyroid-stimulating horestablishing a study design as well as potential mone [84]. Such a thyroidal state is referred to as confounders when interpreting immune function the “euthyroid sick syndrome” and can accomresults. Protein consumption during exercise pany severe body weight loss [3, 52, 84]. There is blunts protein degradation and has a sparing also an effect on the adrenocortical axis, with effect on muscle glycogen [72]. higher levels of cortisol due to an increased liberation of corticotropin-releasing hormone [84]. Growth Phase (Post-exercise) Immediate post-­ Growth hormone is also increased, although workout fuel consumption has the potential to insulin-like growth factor-1 levels (which facilihighly influence muscle machinery by utilizing tate the physiological actions of growth hormone) the anabolic characteristics of insulin. are suppressed in the anorexia condition [84]. Additionally, an increase in insulin post-exercise Due to the psychological aspects of the anorexia post-exercise. Although most research protocols hold diet constant, considerations for what the subject consumes before and after, ad libitum, may have substantial influences on acute and chronic adaptations, in part due to the stimulation of hormones. 1 Methodological Considerations in Exercise Endocrinology nervosa (see Refs. [85, 86]), this condition could, in the context of the organization of this chapter, be also discussed with mental health issues. Thus this factor could also be considered of a biological nature and consequently has powerful effects on a multitude of endocrine measurements. 9 [92, 93]. If inadequate amounts of time have elapsed (lack of recovery), some hormonal responses at rest, or in the subsequent exercise testing, can be attenuated and others augmented. Furthermore, the magnitude of this effect can be influenced by the exertion required of the prior exercise (e.g., high-intensity intervals require longer recovery). Stress-Sleep If possible, researchers may require a 24-h recovery prior to a subject reporting to the laboEmotional stress and/or sleep deprivation are ratory for testing. However, subjects who are each known to affect certain hormones within the athletes may find it difficult to reduce their endocrine system. For example, emotionally dis- training or miss a workout session for experitraught individuals will typically have elevated mental purposes in research studies. A modified basal catecholamine, growth hormone, cortisol, approach may be necessary, such as only a 12and prolactin levels [1, 87–89]. Those hormones or 8-h recovery period, because this could somewith circadian patterns (see Circadian Rhythm what prevent stress and anxiety (which as noted section; e.g., luteinizing hormone, follicle-­ can affect the endocrine system) in the athlete stimulating hormone, adrenocorticotropic hor- since they would be missing less training time mone, cortisol) can be shifted in their [1, 93–95]. characteristic pattern-rhythm by disruption of A powerful influence on resting and exercise sleep cycles [43, 46, 87–91]. hormonal response of a subject is the exercise These types of factors (i.e., stress, sleep depri- training status—that is, trained vs. sedentary. The vation) can also influence the hormonal response more “trained” a subject is, typically the greater to exercise and exercise training. Investigators the effect on the neuroendocrine system. Many must attempt to control these factors whenever hormones show attenuated resting and submaxipossible. In fact, it is advisable to have a pre-­ mal exercise responses in trained individuals, exercise questionnaire completed by a subject to although some can actually be augmented (e.g., monitor and evaluate the level of these factors, testosterone in resistance-trained individuals) in and if a predetermined status is not obtained, then response to submaximal and maximal exercise hormonal measures and exercise testing should [2, 96–100]. be rescheduled. Besides acute effects of physical activity on As a footnote to this issue, many investiga- hormonal levels, chronic endurance exercise tions in the exercise area use college students as such as distance running, cycling, race walking, research subjects. Such students can have high and triathlon has been shown to put immense levels of emotional stress due to their education stress on the endocrine system, many times demands (e.g., examination periods, projects resulting in the suppression of some hormones being due, oral reports). Care should be taken to [9]. This phenomenon may be related to the not utilize student subjects when there are in high “overtraining syndrome” (see Chap. 27 of this emotion stress periods as a multitude of hor- book). The exact mechanism of hormonal reducmones can potentially have very atypical values tion following chronic strenuous endurance and responses [43]. exercise is not elucidated yet; however an impairment of the hypothalamic-­ pituitary regulatory axis due to energy decreases is postulated by Physical Activity some researchers [101]. An extensive dialogue on the influence of The proximity in time between exercise sessions exercise training on hormonal profiles at rest and can affect the hormonal profiles of individuals in response to exercise is beyond the scope of this 10 chapter, but the reader is directed to Refs. [2, 3] for more in-depth discussions. Subject Posture-Position There are changes in the plasma volume component of the blood as a subject changes position. Standing upright results in a reduction of plasma volume compared to a recumbent position [102]. These shifts in the plasma fluid are in response to gravitation effects as well as alterations in capillary filtration and osmotic pressures [102]. Large molecular size hormones, or ones bound to large weight carrier proteins, could be trapped in the vascular spaces; this means that a loss of plasma fluid would increase the concentration of these hormones (hemoconcentration). Conversely, a gain of plasma fluid would decrease the concentration of these hormones (hemodilution) [44, 103]. These adjustments in fluid volume to move in or out of the vascular space due to posture shifts typically require approximately 10–30 min [102, 103]. In exercise research situations where blood is drawn to assess hormones, it is recommended that the condition of specimen collection related to the subject’s position be controlled and reported in publications. This type of information is most certainly necessary if a postural change is occurring for a 10-min or greater duration [58, 103]. Specimen Collection Suitable precautions must be taken in the collection and storage of blood specimens to ensure they are viable for later hormonal analysis. In clinical and exercise-related blood work, venous blood is the specimen usually utilized. If the blood specimen is being obtained by venipuncture, it is important to not have the tourniquet on the subject’s arm too long (∼1 min or longer). Greater lengths of time can result in fluid movement from the vascular bed due to increased hydrostatic pressures [103]. Once collected, the A. C. Hackney et al. blood sample should be centrifuged at ∼ 4 °C in order to separate the plasma (collection tube contains anticoagulant) or allowed to clot (collection tube is sterile) then centrifuged for serum. If centrifugation cannot be done immediately, then the blood sample should be placed on ice, but it is more prudent to centrifuge without delay. Once separated, the plasma/serum should be aliquoted and stored at a temperature of −20 to −80 °C until later analysis. Care should be given to ensure certain plasma/serum is stored in airtight cryofreeze tubes (screw-cap type is recommended), which allow for a longer storage period. It is also advisable to split up specimens into several aliquots if multiple hormonal analyses are going to be conducted. Once a sample is thawed, it has a relatively short “shelf life” in a refrigerator, and repeated unthawing and refreezing cycles can degrade certain hormonal constituents and compromise the validity of the analysis [104– 106]. Care should be taken to ensure that the assay procedures employed are specific for plasma or serum, as in some cases these cannot be used interchangeably in the assay (e.g., adrenocorticotropic hormone is measured in plasma). Furthermore, an examination of the research literature may be necessary to determine if one form of blood component is more popular or prevalently used in research. In blood specimens, either plasma or serum is utilized for biochemical analysis, but some hormonal measures can also be made in urine and salivary samples. In general plasma and serum give very similar values for hormonal analytes, and seldom is one considered better than the other in blood analysis [105, 106]. Be aware, however, specific assay procedures do, in some situations, have a preferred blood fluid for analysis. Thus it is critical for the researcher to know what each hormonal assay requires as the analyte and then plan accordingly. This type of information is provided by the manufacturer of the analytical supplies-components used in the assay procedures. With respect to urine and saliva, they are attractive as specimens to collect because of their noninvasive nature. They do, however, have cer- 1 Methodological Considerations in Exercise Endocrinology tain drawbacks. Urine analysis tends to be limited primarily to steroid-based hormones, and there is usually a need to collect 24-h urine specimen. The collection of 24-h urine specimens can be a tedious and demanding process for the subject. Also, urine measurements may not always be reflective of “real-time” hormonal status either, as urine can sit in the bladder for hours before being voided. Saliva allows for easier sampling procedure and can reflect hormonal status in a more real-time fashion. However, saliva also primarily only allows for steroid hormonal assessments (i.e., constituents that can cross from the blood into the salivary gland) [107]. Furthermore, saliva is limited to free hormonal concentrations as the protein-bound constituents typically cannot pass through the salivary gland due to their large molecular size. Research does suggest that the blood and saliva levels of hormones can mirror each other in their relative changes, but not perfectly, as correlation coefficients of only 0.7–0.8 are typically found [1, 104, 107]. Researchers must determine if these limitations preclude the use of these biological fluids in their studies [104, 107–109]. Analytical Assays A variety of biochemical analytical methods (i.e., “assays”) exist for measuring hormones in biological specimens. Chromatographic, receptor, and immunological assays are all available. Perhaps the most prevalent contemporary technique in use is immunological assays, which have variations such as chemiluminescence immunoassay (CLIA), radioimmunoassays (RIA), enzyme immunoassays (EIA), enzyme-linked immunoassays (ELISA), and electrochemiluminescence immunoassays (ECLIA) [109–111]. Each of these techniques has its strengths and weaknesses, and the discussion of each is beyond the scope of this chapter, but the reader is directed to Refs. [112–114] for more background and explanation about this subject. Researchers should always know the particular aspects of the hormonal assay techniques they 11 plan to use in their studies. Specifically, it is important they be aware of the precision of the assay (“how accurate is it?”), sensitivity of the assay (“how small of a change can it detect?”), and the specificity of the assay (“how much cross-reactivity is there with similar looking chemical structures in the specimen?”). Ideally the researcher wants the most precise, highly sensitive, and specific assay they can obtain, but cost considerations can impact decision-making in these matters. It is advisable for the researcher to report precision, sensitivity, and cross-reactivity values in publications to allow readers to determine the quality of the analytical techniques and procedures of the assays that were used. Additionally, it is desirable to report in publications the coefficient of variation (CV) “within” an assay and “between” an assay for each respective hormone measured. This will allow the reader to determine how well the analytical technical procedures were carried out [114, 115]. One step to mitigate the potential between-assay CV is to collect and analyze your biological samples in batches of specimens and not as isolated specimens on a day-by-day basis. However, caution is necessary here as batches that are too large can influence your outcome by creating “end of run effects” within the assay. That is, running such a large number of samples in a single batch that the precision of the technician performing the assay may be compromised (i.e., procedural fatigue), or the kinetics of the specific assay may be influenced by the length of time it takes to pipette the various components in assay (i.e., in adding the chemical reagents to the first sample tubes vs. the last tubes; too much time has transpired, resulting in different lengths of time for chemical reactions to take place within the specimen tubes) [114, 115]. Data Transformations Before conducting statistical analysis on hormonal data measured within the assays, it may be necessary to transform the data. Two of the most common endocrine transformations usually seen A. C. Hackney et al. 12 in literature are (1) expressing the data as a percent change from some precondition (i.e., before exercise), basal value, and (2) conducting a logarithmic conversion of the data. The first is ­typically done to account for relative changes in hormonal concentrations when absolute magnitude of change may be misleading. For example, a cortisol change from 276 to 331 nmol/L is highly different from a 55–110 nmol/L (20% vs. 200%) even though the absolute magnitude is identical. A 200% increase in the hormonal concentration may have many more profound physiological effects than the smaller percentage. In the second form of transformation, logarithmic transformation is normally performed due to a large degree of variance in the subject data resulting in a nonnormal distribution. This can be due to sample size issues, variance with the analytical technique, or the physiological nature of the hormone being studied. Despite the transformation used, it is vital that the researcher report to the reader in the publication if and how the data were manipulated prior to conducting the statistical analysis (and what was the rationale for performing the transformation) [109, 116]. A third data transformation that is less frequently used is the area under-the-curve (AUC) procedure. This is carried out when there are serial specimen samples (repeated measures design) from a subject. These serial values are plotted, and then an integration of the area under the plotted responses curve is determined, thus collapsing numerous data values into one response and potentially eliminating some of the variability associated with having many hormonal measurements [117]. This approach is favored by some researchers; their rationale is the overall response of the hormone, and gland in question can be better quantified. Nonetheless, the procedure can be influenced by the number of serial samples collected to determine the response curve as well as the circadian rhythm of the hormonal release. The latter point results in the need for highly variable hormones (pulsatile) to be assessed using more frequent specimen sampling because misleading results can occur if the sampling is too infrequent [118]. Statistical Analysis The statistical analytical procedures applied to any research study data are dictated by the design of that study. Most research in the exercise area tends to employee parametric analysis (e.g., t-test, one-way ANOVA, Pearson correlation). These analytical procedures work well with endocrine data, provided that the underlying assumptions for their use are not violated (see Ref. [119] for details). Furthermore, many North American journals prefer this form of analysis due to the robust nature of the techniques and the reduced likelihood of making a type I error (indicating findings are significant when they are in fact, not). Nevertheless, nonparametric analysis (e.g., Wilcoxon signed-rank test, Mann-Whitney U test, Friedman test) can be equally applicable for endocrine use when study designs are not excessively complex and sample sizes are relatively small [119]. It is important, however, to recognize the likelihood of increasing the occurrence of a type I error with small sample sizes. Regardless of whether parametric or nonparametric analyses are used, it is vital that the researcher report in a publication of their work what the specific statistical analysis being used is and what the rationale was for their usage [118–121]. Once assays are performed and statistical results are obtained, the researcher needs to try and understand their data in order to interpret the magnitude of treatment outcomes and physiological effects. In this interpretative process, many researchers focus intently only upon obtaining statistical significance, usually a probability level less than 0.05 (p < 0.05). Obtaining such significance is important; however, a key question that has to be addressed in the data is the issue of “statistical significance” vs. “practical (clinical) significance” for the hormonal findings. To address that question, the researcher must think about and take into account the smallest clinically important positive and negative response value levels of the effect being researched, that is, the smallest change value levels that matter. Studies can be statistically sig- 1 Methodological Considerations in Exercise Endocrinology 13 nificant yet largely insignificant clinically. It is References important to note that large sample sizes can pro1. Trembly MS, Chu SY, Mureika R. Methodological duce a statistically significant result even though and statistical considerations for exercise-related horthere might be limited or no practical importance mone evaluations. Sports Med. 1990;20(2):90–108. associated with the findings [122]. 2. Kraemer WJ, Ratamess NA. Hormonal responses and adaptations to resistance exercise and training. To this end, effect sizes (ES) are becoming an Sports Med. 2005;35(4):339–61. increasingly important index used to quantify the 3. McMurray RG, Hackney AC. The endocrine system degree of practical significance of study results and exercise. In: Garrett W, editor. Sports medicine. (see Ref. [123] for explanation to calculating the New York: Williams & Wilkins; 2000. p. 135–62. 4. International Union of Pure and Applied Chemistry ES statistic). Once computed, the ES statistic can (International Union of Biochemistry and Molecular be a useful indicator of the practical-clinical Biology): recommendations on organic & biochemiimportance of research results because it can be cal nomenclature, symbols & terminology. www. operationally defined; that is, it is possible to give chem.qmul.ac.uk/iupac/. the observed ES ratings such as “negligible-­ 5. Warne GL, Kanumakala S. Molecular endocrinology of sex differentiation. Sem Reprod Med. trivial,” “moderate,” or “important-very large” 2002;20(3):169–80. [124]. Such ratings allow the researcher to dis6. Webb ML, Wallace JP, Hamill C, Hodgson JL, cern the form and quantity of significance they Mashaly MM. Serum testosterone concentration during two hours of moderate intensity treadmill have obtained in their study findings. In addition, running in trained and untrained men and women. the ES statistic has two advantages over tradiEndocrinol Res. 1984;10:27–38. tional statistical significance testing: (a) it is 7. Bunt JC, Bahr JM, Bemben DA. Comparison of independent of the size of the sample, and (b) it is estradiol and testosterone levels during and immediately following prolonged exercise in modera scale-free index. Thus, ES can be uniformly ately active males and females. Endocrinol Res. interpreted in different studies regardless of the 1987;13:157–72. sample size and the original scales of the vari8. Pedersen BK, Hoffman-Goetz L. Exercise and the ables being examined [123, 124]. immune system: regulation, integration, and adapta- Summary and Conclusion To conclude, over the last several decades, exercise researchers have steadily increased the number of studies conducted which have examined the hormones and the endocrine system. Unfortunately, not all investigators working in this area of research are entirely aware of the factors that must be accounted for, and controlled, in order to ensure that valid and accurate data are obtained. This chapter reviewed some of the key physiological and procedural-analytical factors that can confound endocrine data and add variance to hormonal findings and those steps to be taken to reduce the confounding factors. Implementation of these steps can greatly aid the researcher in the interpretation and understanding of their endocrine data and in turn make their research more scientifically sound. tion. Physiol Rev. 2000;80:1055–81. 9. Foster DL, Nagatani S. Physiological perspectives on leptin as a regulator of reproduction: role in timing puberty. Biol Reprod. 1999;60(2):205–12. 10. Ruby BC, Robergs RA. Gender differences in substrate utilization during exercise. Sports Med. 1994;17:393–410. 11. Heavens KR, Szivak TK, Hooper DR, Dunn-­ Lewis C, Comstock BA, Flanagan SD, Looney DP, Kupchak BR, Maresh CM, Volek JS. The effects of high intensity short rest resistance exercise on muscle damage markers in men and women. J Strength Cond Res. 2014;28:1041–9. 12. Bunt JC. Metabolic actions of estradiol: significance for acute and chronic exercise responses. Med Sci Sports Exerc. 1990;22(3):286–90. 13. Hackney AC, McCracken M, Ainsworth BA. Substrate metabolism responses to submaximal exercise in the mid-follicular and mid-luteal phase of the menstrual cycle. Int J Sport Nutr. 1994;4:299–308. 14. Hackney AC, McMurray RG, Judelson DA, Harrell JS. Relationship between caloric intake, body composition, and physical activity to leptin, thyroid hormones, and cortisol in adolescents. Jpn J Physiol. 2003;53(6):475–9. 14 15. Horswill CA, Zipf WB, Kien CL, Kahl EB. Insulin’s contribution to growth in children and the potential for exercise to mediate insulin’s action. Pediatr Exerc Sci. 1997;9:18–32. 16. Amile SA, Caprio S, Sherwin RS, Plewe G, Haymond MW, Tamborlane WV. Insulin resistance of puberty: a defect restricted to peripheral glucose metabolism. J Clin Endocrinol Metab. 1991;72:277–82. 17. Isurugi K, Fukutani K, Takayasu H, Wakabayashi K, Tamaoki B. Age related changes in serum LH and FSH level in normal men. J Clin Endocrinol Metab. 1974;39:955–7. 18. Purifoy EE, Koopmars LH, Tatum RW. Steroid hormones and aging: free testosterone, testosterone and androstenedione in normal females age 20–87 years. Hum Biol. 1980;52:181–91. 19. Orentreich N, Brind JL, Rizer RL, Vogelman JH. Age changes and sex differences in serum dehydroepiandrosterone sulfate concentrations throughout adulthood. J Clin Endocrinol Metab. 1984;59:551–5. 20. Brook MS, Wilkinson DJ, Mitchell WK, Lund JN, Phillips BE, Szewczyk NJ, Greenhaff PL, Smith K, Atherton PJ. Synchronous deficits in cumulative muscle protein synthesis and ribosomal biogenesis underlie age-related anabolic resistance to exercise in humans. J Physiol. 2016;594:7399–417. 21. Aloia JF, Feuerman M, Yeh JK. Reference range for serum parathyroid hormone. Endocr Pract. 2006;12(2):137–44. 22. Adlercreutz H, Goldin BR. Estrogen metabolism and excretion in Oriental and Caucasian women. J Natl Cancer Inst. 1994;86:1076–82. 23. Benn PA, Clive JM, Collins R. Medians for second trimester maternal serum AFP, unconjugated estriol, and hCG: differences between race or ethnic groups. Clin Chem. 1997;43:333–7. 24. Mittelmark RA. Hormonal responses to exercise in pregnancy. In: Mittelmark RA, Wiswell RA, Drinkwater BL, editors. Exercise in pregnancy. Baltimore: Williams & Wilkins; 1991. p. 175–84. 25. Wang C, Christenson P, Swerdloff R. Clinical relevance of racial and ethnic differences in sex steroids. J Clin Endocrinol Metab. 2007;92(7):2433–5. 26. Abbott WG, Foley JE. Comparison of body composition, adipocyte size, and glucose and insulin concentrations in Pima Indian and Caucasian children. Metabolism. 1987;36(6):576–9. 27. Punjani N, Nayan M, Grober E, Lo K, Lau S, Jarvi K. The effect of ethnicity and race on semen analysis and hormones in the infertile patient. J Urol. 2018;199:e248. 28. Fink J, Matsumoto M, Tamura Y. Potential application of testosterone replacement therapy as treatment for obesity and type 2 diabetes in men. Steroids. 2018;138:161–6. 29. Ivandic A, Prpic-Krizevac I, Sucic M. Hyperinsulinemia and sex hormone in healthy premenopausal women: relative contribution of obesity, obese type, and duration of obesity. Metabolism. 1998;47:13–9. A. C. Hackney et al. 30. Hansen BC, Jen KL, Pek SB. Rapid oscillations on plasma insulin, glucagons, and glucose in obese and normal weight humans. J Clin Endocrinol Metab. 1982;54(4):785–92. 31. Florkowski CM, Collier GR, Zimmet PZ. Low dose growth hormone replacement lowers plasma leptin and fat stores without affecting body mass index in adults with growth hormone deficiency. Clin Endocrinol. 1996;45:769–73. 32. Pasquali R, Vicennati V. Activity of the hypothalamic-­ pituitary-adrenal axis in different obese phenotypes. Int J Obes Relat Metab Disord. 2000;24(Suppl 3):S47–9. 33. McMurray RG, Hackney AC. Interactions of metabolic hormones, adipose tissue and exercise. Sports Med. 2005;35(5):393–412. 34. Hurley BF, Nemeth PM, Martin WH. Muscle triglyceride utilization during exercise: effect of training. J Appl Physiol. 1986;60:562–7. 35. Rahkila P, Soimajarvi J, Karvinrn E. Lipid metabolism during exercise II: respiratory exchange ratio and muscle glycogen content during 4 h bicycle ergometry and two groups of health men. Eur J Appl Physiol. 1980;44(3):246–54. 36. Pasman WJ, Westertrep-Plantenga MS, Saris WHM. The effect of exercise training on leptin levels in obese males. Am J Physiol Endocrinol Metab. 1998;37:E280–6. 37. Ryan AS, Partley RE, Elahi D. Changes in leptin and insulin action with resistive training in postmenopausal women. Int J Obes Relat Metab Disord. 2000;24:27–32. 38. Rabkin JG, Wagner GJ, Rabkin R. A double-blind, placebo-controlled trial of testosterone therapy for HIV-positive men with hypogonadal symptoms. Arch Gen Psychiatry. 2000;57:141–7. 39. Grossmann M. Low testosterone in men with type 2 diabetes: significance and treatment. J Clin Endocrinol Metab. 2011;96:2341–53. 40. Wong N, Levy M, Stephenson I. Hypogonadism in the HIV-infected man. Curr Treat Options Infect Dis. 2017;9:104–16. 41. Hackney AC. Stress and the neuroendocrine system: the role of exercise as a stressor and modifier of stress. Expert Rev Endocrinol Metab. 2006;1(6): 783–92. 42. Dorn LD, Burgress ES, Dichek HL, Putman FW, Chrousos GP, Gold PW. Thyroid hormone concentrations in depressed and nondepressed adolescents: group difference and behavioral relations. J Am Acad Child Adolesc Psychiatry. 1996;35:299–306. 43. Vaernes R, Ursin H, Darragh A, Lambe R. Endocrine response patterns and psychological correlates. J Psychosom Res. 1982;26:123–31. 44. Hackney AC. Exercise as a stressor to the neuroendocrine system. Medicina. 2006;42(10):788–97. 45. Hammer MB, Hitri A. Plasma β-endorphin levels in post-traumatic stress disorder: a preliminary report on response to exercise-induced stress. J Neuropsychiatry Clin Neurosci. 1992;4(1):59–63. 1 Methodological Considerations in Exercise Endocrinology 46. Gerra G, Volpi R, Delsignore R, et al. ACTH and β-endorphin responses to physical exercise in adolescent women tested for anxiety and frustration. Psychiatry Res. 1992;41(2):179–86. 47. Cohen S, Kamarck T, Mermelstein R. A global measure of perceived stress. J Health Soc Behav. 1983;24(4):385–96. 48. Beck AT, Epstein N, Brown G, Steer RA. An inventory for measuring clinical anxiety: psychometric properties. J Consult Clin Psychol. 1988;56(6):893–7. 49. Landgren B, Aedo A, Diczfalusy E. Hormonal changes associated with ovulation and luteal function. In: Flamigni C, Givens J, editors. The gonadotropins: basic science and clinical aspects in females. London: Academic; 1982. p. 200–12. 50. Hackney AC, Cyren HC, Brammeier M, Sharp RL. Effects of the menstrual cycle on insulin-­ glucose at rest and in response to exercise. Biol Sport. 1993;10(2):73–81. 51. Vanheest JL, Mahoney CE, Rodgers CD. Oral contraceptive use and physical performance. In: Kraemer WJ, Rogol A, editors. The endocrine system in sports and exercise. Oxford: Blackwell; 2005. p. 250–60. 52. Loucks AB. Physical activity, fitness and female reproductive morbidity. In: Bouchard C, Shepard RJ, Stephens T, editors. Physical activity, fitness and health: international proceedings and consensus statement. Champaign: Human Kinetics; 1994. p. 943–54. 53. Matsumoto AM, Bremner WJ. Modulation of pulsatile gonadotropin secretion by testosterone in man. J Clin Endocrinol Metab. 1984;58(4):609–14. 54. Rose R, Kreutz L, Holoday J, Sulak K, Johnson C. Diurnal variation of plasma testosterone and cortisol. J Endocrinol. 1972;54:177–8. 55. Rose SR, Nisula BC. Circadian variation of thyrotropin in childhood. J Clin Endocrinol Metab. 1989;68:1086–9. 56. Hackney AC, Viru A. Twenty-four cortisol response to multiple daily exercise sessions of moderate and high intensity. Clin Physiol. 1999;19:178–82. 57. Weitzman ED. Circadian rhythms and episodic hormone secretion. Annu Rev Med. 1976;27:225–43. 58. Goodman HM. Endocrinology concepts for medical students. Adv Physiol Educ. 2005;25(4):213–24. 59. Hackney AC, Zack E. Physiological day-to-day variability of select hormones at rest in exercise-trained men. J Endocrinol Investig. 2006;29(6):RC9–12. 60. Schulz P, Knabe R. Biological uniqueness and the definition of normality: part 2—the endocrine ‘finger print’ of healthy adults. Med Hypotheses. 1994;42:63–8. 61. Antonio L, Wu FC, O'neill TW, Pye SR, Ahern TB, Laurent MR, Huhtaniemi IT, Lean ME, Keevil BG, Rastrelli G. Low free testosterone is associated with hypogonadal signs and symptoms in men with normal total testosterone. J Clin Endocrinol Metabol. 2016;101:2647–57. 15 62. Finberg JP, Berlyne GM. Renin and aldosterone secretion following acute environmental heat exposure. Isr J Med Sci. 1976;12:844–7. 63. Galbo H, Houston ME, Christensen NJ, Holst JJ, Nielsen B, Nygaard E, et al. The effect of water temperature on the hormonal response to prolonged swimming. Acta Physiol Scand. 1979;105(3):326–37. 64. Mordes JP, Blume FD, Boyer S, Zheng MR, Braverman LE. High altitude pituitary-thyroid dysfunction on Mount Everest. N Engl J Med. 1983;308:1135–8. 65. Rastogi GK, Malhotra MS, Srivastava MC, Shawhney RC, Dua GL, Sridharan K, et al. Study of the pituitary-thyroid function at high altitude in man. J Clin Endocrinol Metab. 1977;43:447–52. 66. Hoyt RW, Honig A. Body fluid and energy metabolism at high altitude. In: Fregley MJ, Blatteis CM, editors. Handbook of physiology, section 4: environmental physiology. New York: Oxford University Press; 1996. p. 1277–89. 67. Galbo H, Holst JJ, Christensen NJ. The effect of different diets and of insulin on the hormonal response to prolonged exercise. Acta Physiol Scand. 1979;107(1):19–32. 68. Phinney SD, Horton ES, Sims EA, Hanson JS, Danforth E, LaGrange BM. Capacity for moderate exercise in obese subjects after adaptation to a hypocaloric, ketogenic diet. J Clin Invest. 1980;66(5):1152–61. 69. Jezova-Repcekova D, Vigas M, Klimes I. Decreased plasma cortisol response to pharmacological stimuli after glucose load in man. Endocrinol Exp. 1980;14(2):113–20. 70. Bonen A, Belcastro AN, MacIntyre K, Gardner J. Hormonal responses during intense exercise preceded by glucose ingestion. Can J Appl Sport Sci. 1980;5(2):85–90. 71. Ivy J, Portman R. Nutrient timing system: the revolutionary new system that adds the missing dimension to sports nutrition: the dimension of time. North Bergen: Basic Health; 2004. p. 33–67. 72. Kerksick C, Harvey T, Stout J, Campbell B, Wilborn C, Kreider R, et al. International Society of Sports Nutrition position stand: nutrient timing. J Int Soc Sports Nutr. 2008;5:17–29. 73. Bishop NC, Blannin AK, Robson PJ, Walsh NP, Gleeson M. The effects of carbohydrate supplementation on immune responses to a soccer-specific exercise protocol. J Sports Sci. 1999;17(10):787–96. 74. Bishop NC, Gleeson M, Nicholas CW, Ali A. Influence of carbohydrate supplementation on plasma cytokine and neutrophil degranulation responses to high intensity intermittent exercise. Int J Sport Nutr Exerc Metab. 2002;12(2):145–56. 75. Lancaster GI, Jentjens RL, Moseley L, Jeukendrup AE, Gleeson M. Effect of pre-exercise carbohydrate ingestion on plasma cytokine, stress hormone, and neutrophil degranulation responses to ­continuous, A. C. Hackney et al. 16 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. high-intensity exercise. Int J Sport Nutr Exerc Metab. 2003;13(4):436–53. Rokitzki L, Logemann E, Huber G, Keck E, Keul J. Alpha-Tocopherol supplementation in racing cyclists during extreme endurance training. Int J Sport Nutr. 1994;4(3):253–64. Nieman DC. Influence of carbohydrate on the immune response to intensive, prolonged exercise. Exerc Immunol Rev. 1998;4:64–76. Nieman DC, Davis JM, Henson DA, Walberg-­ Rankin J, Shute M, Dumke CL, et al. Carbohydrate ingestion influences skeletal muscle cytokine mRNA and plasma cytokine levels after a 3-h run. J Appl Physiol. 2003;94(5):1917–25. Tipton KD, Rasmussen BB, Miller SL, Wolf SE, Owens-Stovall SK, Petrini BE, et al. Timing of amino acid-carbohydrate ingestion alters anabolic response of muscle to resistance exercise. Am J Physiol Endocrinol Metab. 2001;281(2): E197–206. Hsu MC, Chien KY, Hsu CC, Chung CJ, Chan KH, Su B. Effects of BCAA, arginine and carbohydrate combined drink on post-exercise biochemical response and psychological condition. Chin J Physiol. 2011;54(2):71–8. Betts JA, Beelen M, Stokes KA, Saris WH, van Loon LJ. Endocrine responses during overnight recovery from exercise: impact of nutrition and relationships with muscle protein synthesis. Int J Sport Nutr Exerc Metab. 2011;21(5):398–409. La Bounty PM, Campbell BI, Wilson J, Galvan E, Berardi J, Kleiner SM, et al. International Society of Sports Nutrition position stand: meal frequency. J Int Soc Sports Nutr. 2011;8:4. Schwarz NA, Rigby BR, La Bounty P, Shelmadine B, Bowden RG. A review of weight control strategies and their effects on the regulation of hormonal balance. J Nutr Metab. 2011;2011:237932. Støving RK, Hangaard J, Hansen-Nord M, Hagen C. A review of endocrine changes in anorexia nervosa. J Psychiatr Res. 1999;33(2):139–52. Casper RC. Recognizing eating disorders in women. Psychopharmacol Bull. 1998;34(3):267–9. Södersten P, Bergh C, Zandian M. Psychoneuroendocrinology of anorexia nervosa. Psychoneuroendocrinology. 2006;31(10):1149–53. VanHelder T, Radomski MW. Sleep deprivation and the effect on exercise performance. Sports Med. 1989;7:235–47. Aakvaag A, Bentdal O, Quigstad K, Walstad P, Ronningen H, Fonnum F. Testosterone and testosterone binding globulin (TeBG) in young men during prolonged stress. Int J Androl. 1978;1:22–31. Aakvaag A, Sand T, Opstad PO, Fonnum F. Hormonal changes in serum in young men during prolonged physical strain. Eur J Appl Physiol. 1978;39:283–91. Diamond P, Brisson GR, Candas B, Peronnet F. Trait anxiety, submaximal physical exercise and blood androgens. Eur J Appl Physiol. 1989;58:699–704. 91. Hackney AC, Feith S, Pozos R, Seale J. Effects of high altitude and cold exposure on resting thyroid hormone concentrations. Aviat Space Environ Med. 1995;66:325–9. 92. Viru A, Hackney AC, Valja E, Karelson K, Janson T, Viru M. Influence of prolonged continuous exercise on hormonal responses to subsequent intensive exercise. Eur J Appl Physiol. 2001;85:578–85. 93. Hackney AC. The neuro-endocrine system, overload training, and regeneration. In: Lehmann M, editor. Ulm international conference proceeding: performance, overload training and regeneration. London: Plenum; 1999. p. 173–86. 94. Viru A, Karelson K, Smirnova T. Stability and variability in hormonal responses to prolonged exercise. Int J Sports Med. 1992;13:230–5. 95. Hartley LH, Mason JW, Hogan RP, Jones LG, Kotchen TA, Mougey EH, et al. Multiple hormonal responses to graded exercise in relation to physical training. J Appl Physiol. 1972;33(5):602–6. 96. Richter EA, Sutton JR. Hormonal adaptation to physical activity. In: Bouchard C, Shephard RJ, Stephen T, editors. Physical activity, fitness and health: international proceedings and consensus statement. Champaign: Human Kinetics; 1994. p. 331–42. 97. Luger A, Deuster PA, Kyle SB, Gallucci WT, Montgomery LC, Gold PW. Acute hypothalamic-­ pituitary-­adrenal responses to the stress of treadmill exercise: physiologic adaptations to physical training. N Engl J Med. 1987;316:1309–15. 98. Hackney AC, Sinning WE, Brout BC. Comparison of resting reproductive hormonal profiles in endurance trained and untrained men. Med Sci Sports Exerc. 1988;20(1):60–5. 99. Remes K, Kuoppasalmi K, Adlercreutz H. Effect of long-term physical training on plasma testosterone, androstenedione, luteinizing hormone and sex-­ hormone binding globulin capacity. Sacnd J Clin Lab Invest. 1979;39:743–9. 100. Hakkinen K, Pakarinen A. Acute hormonal responses to two different fatiguing heavy-resistance protocols in male athletes. J Appl Physiol. 1993;74:882–7. 101. Hackney AC, Aggon E. Chronic low testosterone levels in endurance trained men: the exercise-­ hypogonadal male condition. J Biochem Physiol. 2018;1(1):pii: 103. 102. Westendorp RG, Roos AN, Riley LC, Walma S, Frolich M, Mienders AE. Chronic stimulation of atrial natriuretic peptide attenuates the secretory responses to postural changes. Am J Med Sci. 1993;306:371–5. 103. Fawcett JK, Wynn V. Effects of posture on plasma volume and some blood constituents. J Clin Pathol. 1960;13:304–13. 104. Chen YM, Cintron NM, Whitson PA. Long term storage of salivary cortisol samples at room temperature. Clin Chem. 1992;38:304. 105. Calam RR. Reviewing the importance of specimen collection. J Am Med Technol. 1977;38:297–300. 1 Methodological Considerations in Exercise Endocrinology 106. Sanntag O. Hemolysis as interference factor in clinical chemistry. J Clin Chem Clin Biochem. 1986;24:575–7. 107. Obminski Z, Klusiewicz A, Stupnicki R. Changes in salivary and serum cortisol concentrations in junior athletes following exercises of different intensities. Biol Sport. 1994;11:49–57. 108. Caraway WT. Chemical and diagnostic specificity of laboratory tests. Am J Clin Pathol. 1961;37:445–64. 109. Kopchick JJ, Sackman-Sala L, Ding J. Primer: molecular tools used for the understanding of endocrinology. Nat Clin Pract Endocrinol Metab. 2007;3(4):355–68. 110. Bowers LD. Analytical advances in detection of performance enhancing compounds. Clin Chem. 1997;43:1299–304. 111. Dudley RF. Chemiluminescence immunoassay: an alternative to RIA. Lab Med. 1990;21:216–22. 112. Shah VP, Midha KK, Findlay JWA, Hill HM, Hulse JD, McGilveray IJ, et al. Bioanalytic method validation - a revisit with a decade of progress. Pharm Res. 2000;17:1551–7. 113. DeRonde W, Van Der Schouw YT, Pols HAP, Gooren LJG, Muller M, Grobbee DE, et al. Calculation of bioavailable and free testosterone in men: a comparison of 5 published algorithms. Clin Chem. 2006;52(9):1777–84. 114. Rosner W, Auchus RJ, Azziz R, Sluss PM, Raff H. Position statement: utility, limitations and pitfalls in measuring testosterone: an Endocrine Society position statement. J Clin Endocrinol Metab. 2007;92:405–13. 17 115. Rodbard D. Statistical quality control and routine data processing for radioimmunoassay and immunoradiometric assays. Clin Chem. 1974;20(10):1255–70. 116. Fraser CG, Harris EK. Generation and application of data on biological variation in clinical chemistry. Crit Rev Clin Lab Sci. 1989;27:409–37. 117. Hackney AC, Premo MC, McMurray RG. Influence of aerobic versus anaerobic exercise on the relationship between reproductive hormones in men. J Sports Sci. 1995;13(4):305–11. 118. Veldhuis JD, Johnson ML. Deconvolution analysis of hormone data. Methods Enzymol. 1992;210: 539–75. 119. Kingle RD, Johnson GF. Statistical procedures. In: Tietz NW, editor. Textbook of clinical chemistry. Philadelphia: Saunders; 1986. p. 287–355. 120. Pincus SM, Hartman ML, Roelfsema F, Thorner MO, Veldhuis JD. Hormone pulsatility discrimination via course and short time sampling. Am J Physiol Endocrinol Metab. 1999;277:E948–57. 121. Matthews DR. Time series analysis in endocrinology. Acta Paediatr Scand Suppl. 1988;347:55–62. 122. Hopkins WG. Measures of reliability in sports medicine and science. Sports Med. 2000;30(1):1–15. 123. Mohammadreza H, Xu G. A visitor’s guide to effect sizes—statistical significance versus practical (clinical) importance of research findings. Adv Health Sci Educ Theory Pract. 2004;9(3):1573–7. 124. Cohen J. Statistical power analysis for the behavioral sciences. 2nd ed. Englewood: Lawrence Erlbaum; 1988. p. 116–73. 2 Endogenous Opiates and Exercise-­ Related Hypoalgesia Allan H. Goldfarb, Robert R. Kraemer, and Brandon A. Baiamonte Introduction (Endogenous Opiates) Endogenous opiate-like substances were first discovered in the mid-1970s, when opioid receptors were identified and located within the brain and hypothalamus [135]. This led to the discovery that endogenous opioid-like molecules, enkephalins [69] and endorphins [9, 106], were produced within the CNS. Subsequently another class of opiate-like molecules known as dynorphins was identified within the body [14, 50]. The latest addition are nociception/orphanin FQ molecules which work on nociceptin opioid receptors (NOP) within the CNS and counteracts the analgesic effect of opiates. The endogenous opiates fall into four major classes of substances: endorphins, a peptide 31 amino acids in length; enkephalins, smaller peptide molecules that are five amino acids in length (denoted either as leuor met-, based on the terminal carboxyl amino acid of the peptide); dynorphins, located in the A. H. Goldfarb (*) University of North Carolina Greensboro, Department of Kinesiology, Greensboro, NC, USA e-mail: ahgoldfa@uncg.edu R. R. Kraemer Southeastern Louisiana University, Department of Kinesiology and Health Studies, Hammond, LA, USA e-mail: rkraemer@selu.edu B. A. Baiamonte Southeastern Louisiana University, Department of Psychology, Hammond, LA, USA e-mail: brandon.baiamonte@selu.edu posterior lobe of the pituitary gland [86, 107] and gastrointestinal tract [60] with a 13 amino acid length; and nociception/orphanin FQ molecules, a peptide of 17 amino acids, which binds to NOP receptors. Enkephalins were first noted in areas of the brain and parts of the endocrine system. The original studies noted that both endorphins and enkephalins were important regulators of pain [4, 106]. However, more recent studies have determined that enkephalins not only play an important role with pain regulation but affect cardiac function, cellular growth, immunity, ischemic tolerance, and certain behaviors. Various tissues (heart, smooth and skeletal muscle, kidney, and intestines) in animals and humans have recently been shown to have proenkephalin expression [26]. Recently, inflammatory cells were shown to produce and release these opiates, and endorphins seem to be involved not only in immune function [79, 81, 123], pain modulation [152], and the exercise pressor response [72, 125, 162] but also in metabolic control [71, 80, 110, 111, 171]. Therefore, numerous challenges remain to be clarified concerning the role of these endogenous opiates on these processes as they relate to exercise. This is especially true regarding the control of cellular functions not only under normal conditions but when acute and chronic exercise stress is imposed. Beta-endorphins (βE) were first identified within specific brain regions and the ­hypothalamus and were found to bind to mu-opioid receptors © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_2 19 20 A. H. Goldfarb et al. (MOR). When MOR are activated there is a Peripheral agonists that do not cross the blood strong inhibition of acute pain [175]. βE within brain barrier can produce analgesia through the the circulation was first ascribed to βE release DRG [164]. However, research which used from the anterior pituitary gland after being acti- knockout deletion of mu receptors and DRG vated by factors within the hypothalamus. These nociceptors in the periphery but with intact CNS factors activate the anterior pituitary gland to receptors reported these peripheral receptors synthesize the parent molecule pro-­were not obligatory for analgesia [23]. It was opiomelanocortin (PMOC) which can be cleaved suggested that these receptors could be involved into various active components, one of them with adverse side effects related to tolerance and being βE. POMC is also expressed in the arcuate opioid-induced hyperalgesia (OIH) with chronic nucleus as well as the nucleus of the solitary tract agonist treatments. However, brain regions can within the CNS. βE is therefore an important also contribute to both tolerance and OIH. These neurotransmitter within the brain and a neurohor- peripheral sensory processes appear to activate mone outside the CNS when released into the important aspects to initiate or modulate CNS circulation, to act on mu receptors on target tis- pain circuits [82] and may be activated with exersues throughout the body. cise [21, 22, 91]. The molecule POMC, the precursor polypepIn addition, there is evidence that serotonin tide for several factors that arise from the hypo- release which alters behavior is modified by actithalamus and the paraventricular nucleus (PVN) vated opioid receptors to influence γ-aminobutyric in the brain, can be stimulated by truncated active acid (GABA) involvement [38]. Additionally, peptides. POMC has a section toward the C ter- interactions have been suggested with MOR with minus known as β-lipotropin (1–89 amino acids) serotonergic structures involved with both reupthat is ultimately cleaved to β-lipotropin take and release of serotonin [159]. Furthermore, (1–56 amino acids) and βE (59–89 amino acids). numerous non-opioid analgesics may influence Both βE and β-lipotropin molecules help to both acute and chronic pain stress [59] and some mobilize lipid molecules from adipose tissue. are related to cannabinoid action [10, 24, 53, 67, Originally the assays that were developed to 161]. Therefore, caution should be taken when measure these molecules did not effectively dif- considering the interpretation of changes which ferentiate between β-lipotropin and βE, which only measure opioid-like agents without assesswere thus denoted as having both β-lipotropin/βE ing alternative pain influencing agents. There are activities. many circuits that can influence pain or its Neuroanatomical sites for opioid analgesia are attenuation. present within the CNS and located on neurons There is limited information related to exerwithin the dorsal root ganglia (DRG) originating cise and brain βE modulation [66, 138, 149]. βE from peripheral somatosensory DRG neurons immunoactivity in cerebrospinal fluid (CSF) of that can transmit these activated processes spontaneously hypertensive rats was shown to be through the spinal cord to the medulla [5]. It significantly higher (about twofold) in runners should be noted that opioid receptors are (5–6 weeks) than in controls [66]. This study expressed on these somatosensory neurons pass- also reported that CSF βE was elevated up to ing through the DRG and have been reported to 48 hours after cessation of voluntary wheel runhave the ability to inhibit or reduce pain percep- ning. It was suggested that this βE effect may be tion [16]. Both mu and delta opioid receptors are at least partially responsible for the beneficial located on DRG neurons, and when opioids are effect of exercise on controlling blood pressure activated, a depressed neuropeptide release from [66]. βE immunoactivity taken from CSF in dogs these afferents to CNS neurons occurs. Recent was shown to increase with low-intensity exerresearch has suggested that myelinated mechano- cise but not with high-intensity exercise [138]. sensory neurons appear to regulate DRG hyper- In contrast, circulating βE immunoactivity sensitivity and chronic inflammatory pain [4]. increased in these dogs at both intensities of 2 Endogenous Opiates and Exercise-Related Hypoalgesia exercise [138]. This indicates that the βE level within the brain is not reflected by the amount of βE within the circulation. Rat brain receptor binding of [3H]diprenorphine, a βE analog, was not significantly elevated 1 hour following a swim but was increased in several brain regions (5 of 6) 2 hours after exercise [149]. It is unclear if this was related to changes in βE concentration or a change in receptor availability. Pain threshold increase that occurred with exercise was abolished when naloxone (a receptor antagonist for βE) was injected into brain ventricles after 5 weeks of exercise training [155]. This suggests that the opioids were involved in elevating pain threshold in response to exercise training in these rats. Clearly more work is needed in this area. Specific brain areas that might be involved with BE and pain regulation in response to different types of exercise still needs further investigation. βE within the circulation has been implicated in a number of processes including immune function, pain modulation, and assisting in glucose and lipid homeostasis. The major function of these endogenous opiate-like molecules was first identified as modulators of pain and euphoria based on the receptors they activated. As a result of this, the phenomena known as “runner’s high,” “second wind,” and “exercise dependency” were postulated to be related to this endogenous activity. This chapter will summarize what is currently known about the stimulation of these endogenous opiates in response to exercise or physical activity, and how exercise may induce exercise-­ induced hypoalgesia (EIH). The influence of an acute bout of exercise on the βE response will be presented first as these studies were the impetus of the original research. The influence of training on βE will then be discussed. Then the influence of an acute bout of exercise on enkephalins will be presented followed by training influences on enkephalins. The physiological mechanisms responsible for activation and secretion of these substances will be briefly discussed when known and related to functional outcomes when possible. Finally the effects of EIH will then be presented. 21 I nfluence of Acute Exercise on β-Endorphin Levels The initial studies that were conducted to examine the impact of exercise on endogenous βE levels utilized various modes of exercise. The original articles examined various activities such as running at various distances to determine if blood βE level was elevated [15, 21, 25, 170]. These studies noted elevated βE after the exercise activities which led to more controlled experiments utilizing incremental graded exercise tests in laboratories to ascertain the βE response [48, 61, 62, 120, 130, 139]. These studies suggested that blood βE can increase from 1.5- to 7-fold following these graded exercise tests. The large variation in the βE response was in part attributed to procedural methods for the exercise tests as well as methods to determine βE and possibly related to the subjects utilized. erobic Exercise at Work Intensities A Related to Percentage of VO2 Max on β-Endorphin Several studies determined whether there was an exercise intensity effect on blood βE level. McMurray et al. [118] was one of the first researchers to examine the βE response to a specific exercise intensity. Donevan and Andrew [28] noted that βE did not increase after 8 minutes of cycling at 25% and 50% maximal oxygen uptake (VO2 max) but increased after 75% VO2 max after similar duration. They also reported a greater increase in βE at 95% VO2 max. Goldfarb et al., in that same year, examined the effects of cycling at several intensities of exercise (60%, 70%, and 80% VO2 max) to determine if there was a critical exercise intensity needed to induce circulating βE [46]. βE concentration increased in the two higher exercise intensities but not at 60% VO2 max. The time course of βE changes at these exercise intensities up to 30 minutes of exercise was examined with βE increases occurring earlier with the highest exercise intensity (by 5 minutes). Research comparing 60% VO2 max and 80% VO2 max as well as self-paced running 22 for 30 minutes noted only an increase after the 80% run [33]; however, they utilized βE/Blipotropin immunoreactivity. A run at 60% VO2 max for 60 minutes induced no change in βE [103]. Exercise at 80% VO2 max for 30 minutes with or without naloxone increased βE with a greater augmented increase with naloxone [1]. These studies taken together suggested that circulating βE increases with an appropriate minimal exercise intensity (>60% VO2 max), but this was not always the case. The time course information also suggested that higher intensities of exercise would result in βE increases more rapidly [33, 35, 49, 61]. Later it was reported that gender did not influence the βE response to either 60% or 80% VO2 max [32, 61, 65, 140]. It was noted that menstrual cycle had minimal effects on the exercise βE response in women [45, 49]. However, other factors might have differed which could have contributed to the discrepancy in the literature such as nutritional status of the individuals, time of day, immune function, and training status. Farrell et al. noted that βE+/β-lipotropin levels in well-trained endurance athletes only increased at 92% VO2 max whereas lower intensities did not elicit significant increases [33]. Instead of a critical intensity relative to one’s maximal aerobic capacity, other studies related the increase in circulating βE to lactate threshold [148]. They plotted the change in lactate with increased work intensity and compared the βE response. Incremental increases in exercise intensity elevated circulating βE levels and showed a similar pattern of change as blood lactate. However, it should be noted that these similar changes are only for shortduration incremental exercise. For activities with longer duration, the βE increase does not coincide with lactate changes [46]. In addition, other factors such as diet, training status, and immune function can influence the βE response. High-Intensity Bouts with an Anaerobic Component Short bouts of highly intensive exercise (anaerobic exercise), consisting of various types of exercise from a few seconds up to several minutes A. H. Goldfarb et al. duration, can induce an increase of βE. A few studies reported that βE concentration in the circulation can increase about 2–4-fold above resting with these high-intensity anaerobic exercise bouts [35, 120, 139, 148]. Schwarz et al. noted a significant increase in blood catecholamines that correlated with the maximal lactate concentrations in response to exercise. Stimulation of the HPA axis through sympathetic activation appears to be related to the release of βE into the circulation. Investigations of resistance exercise as a stimulus to augment circulating βE concentration in humans includes a limited number of published studies. Equivocal results have been reported, and this may be related to differences in subjects, type of exercise intensity, workload volume, and time of measurement. Typically the resistance exercise was related to the person’s 1-repetition maximum (1-RM), i.e., maximum weight that was lifted or pushed/pulled by a subject with maximal effort. Often the load is referenced as a percentage of the 1-RM. Circulating βE level increased in response to high total workloads [97]. These authors suggested that the total work, rest to work ratio, and total force needed most likely influenced the βE response. An increase in βE in 28 elite male weight lifters was demonstrated after a moderate- to high-intensity workload [98]. An increase in βE level also occurred after three sets of work at 85% 1-RM in females but was significantly elevated (3.7-fold) only when these women were in a negative energy balance [172]. An increased βE/β lipotropin level was reported in response to weight lifting in five males [31]. In contrast, Kraemer et al. conducted a study using low-volume resistance exercise as a stimulus and reported no change in βE levels [95]. Furthermore, blood βE level based on immunoreactivity decreased after exercise compared to at rest in ten male and ten female college-aged ­students who performed three sets of eight repetitions at 80% 1-RM on four exercises [136]. This same group had reported earlier that resistance-­trained subjects (N = 6) showed no change in blood βE level compared to baseline after three sets of eight repetitions at 80% 1-RM [137]. Both resistance exercise and treadmill 2 Endogenous Opiates and Exercise-Related Hypoalgesia exercise were reported to significantly increase circulating βE/B-lipotropin immunoreactivity [31]. Unfortunately the intensity and volume of exercise was not available. McGowan et al., however, noted a decrease in βE concentration after exercise at 80% 1-RM in 20 college-aged subjects (both genders) [117]. It appears that resistance exercise of sufficient intensity and volume (workload) can result in a transient βE increases within the circulation in both men and women, but this finding is sometimes equivocal. Influence of Training on β-Endorphin Levels The training status of the individual can influence the response to exercise for a number of reasons. One reason is related to the relative intensity of the exercise. Well-trained athletes can typically perform at a greater absolute workload and usually would exercise at a higher relative workload compared to an untrained individual. Therefore, when comparing the βE response one should compare the absolute workload and the relative intensity. In addition, other factors might influence the secretion of βE such as the diet or immune function which can be influenced by training. Typically one would expect a downregulation on the secretion of βE to a similar absolute workload. However, there could be an upregulation of the capacity of the hypothalamic–pituitary–adrenal (HPA) axis in trained individuals. Finally, the amount of free hormone and the number of binding receptors could be modulated to influence action on target tissues. Influence of Endurance Training Resting levels of βE in endurance-trained individuals were reported to be lower [66] with the vast amount of studies reporting no change [45, 61, 62, 68]. The studies that reported no changes were mostly cross-sectional studies. In contrast, the study that reported lower levels used an endurance training program and compared the βE level before and after the training program at rest [109]. In contrast, Heitkamp reported that 23 women who trained three times per week for 30 minutes each time at their individualized lactate threshold did not have changes in their resting βE [61]. Harber and associates compared normal eumenorrheic sedentary to eumenorrheic-trained and amenorrheic-­ trained women and reported that βE varied considerably, but there was no menstrual cycle effect at rest on βE [56]. They also noted that resting βE levels were higher in the trained women compared to the sedentary women. Goldfarb et al. reported a trend for lower βE levels during the luteal phase of the menstrual cycle compared with the follicular phase, but this did not reach significance [49]. They also noted no significant difference in βE at rest between men and women. Therefore, there is currently no consensus in the literature as to the effect of endurance training on resting βE levels. The βE response during exercise is in slightly better agreement when exercise intensities were controlled. One early study reported a higher βE concentration after 4 months of aerobic training six times per week [15]. They reported that the βE level was higher cycling at 85% max heart rate (HR) than before training. This occurred after 2 months of training with no further changes. It should be noted, however, that to elicit a similar 85% max HR, the subjects worked at a greater absolute workload. Most of the other studies have reported no detectable differences in trained and untrained subjects regardless of whether it was a cross-­ sectional design [47] or longitudinal design [12, 32, 61, 62, 68]. Goldfarb et al. compared untrained (N = 6) and trained cyclists (N = 6) that cycled for 30 minutes at 60%, 70%, and 80% VO2 max with subjects randomly assigned in a ­counterbalanced order [47]. There was no difference in the βE concentration for the trained and untrained at similar relative workloads despite higher absolute workloads for the trained group. Both untrained and trained groups responded with higher βE levels at both 70% and 80% workloads compared to rest and the 60% workload. Heitkamp et al. reported that after training the βE response was comparable but was obtained at higher absolute workload for the trained subjects [61]. They also reported that after training the 24 recovery βE was lower suggesting faster removal of βE. Howlett et al. also reported no difference in βE concentration after endurance training at maximal workloads but met-enkephalin concentration was reduced after 4 months of training [68]. Bullen et al. reported greater peak βE/β-­lipotropin post-exercise after 8 weeks of cycling training in seven women [12]. Engfred reported similar βE increases after 5 weeks of cycling training at 70% VO2 max cycling to exhaustion [32]. VO2 max increased 12% following 5 weeks of training so a higher absolute workload after training was utilized. In conclusion, it appears that blood βE concentration in trained individuals will be similar to concentrations before training if the workload is at the same relative intensity of aerobic capacity. This would require a higher absolute workload for the trained individual. A. H. Goldfarb et al. pose tissue, pancreas, and skeletal muscle. However, the exact role(s) βE may have on these tissues is still being elucidated. The influence of βE on immune function has been investigated in vitro but has not been adequately investigated in vivo. βE in rats and humans was shown to stimulate T lymphocyte proliferation [63]. The data suggests that βE mode of action was not though a MOR. It was shown that synthetic βE could bind to non-opioid receptors on T lymphocytes, and this binding was not blocked by naloxone or met-enkephalin [126]. In vitro βE stimulated rat spleen lymphocytes in a dose-dependent manner by enhancing the proliferative response to several mitogens [44]. This binding was not blocked by naloxone. βE enhanced the proliferative response of splenocytes on T-cells from adult male F344 rats [165]. In addition, naloxone was not effective in blocking the βE effect. βE stimulated the proliferative Influence of Resistance Training effect on human T lymphocytes using the mitoon Circulating β-Endorphin gen concanavalin A [131]. This βE-stimulated Unfortunately there are few studies that have mitogen response demonstrated a bell-shaped examined the influence of resistance training on curve indicating that too high a dose would actucirculating βE. There are no published studies ally inhibit the response. It was suggested that found which indicate that βE concentration this response may change with time, dose, or would change at rest or at any specific workload mitogen used [121]. These authors also reported or a percentage (%) of one’s maximal capacity that the inhibition of the immune response to maybe partially reversed by with resistance training. Fry and coworkers cortisol reported similar βE concentration after both 4 βE. Therefore, the activation of βE may inhibit and 9 weeks of resistance training to baseline lev- suppression of the immune response by acting els [40]. It is important to note that most of the on cortisol actions in vivo. The βE effect on to enhance natural killer resistance research typically utilized resistance-­ trained subjects. As noted above, higher total (NK) cell function in vitro was reported to be a work volume with resistance exercise resulted in dose-dependent manner but was inhibited by naloxone [85]. This suggests that the mode of greater increases in circulating βE [97]. action on NK cells appears to be different than the enhancement of T lymphocyte function. The effect of βE concentration on NK cell activity β-Endorphin and the Immune (NKCA) and amount was examined after exerSystem cise [43]. Naltrexone treatment administered βE within the circulation has been implicated in a 60 minutes before a run at 65% VO2 max which number of processes including modulation of elevated blood βE levels at 90 and 120 minutes immune function, pain modulation, blood pres- did not alter the exercise response in NKCA or sure regulation, and assisting in glucose homeo- amount. These authors suggested that βE may stasis. βE receptors have been identified in many work independent of the MOR action to assist locations within the body including nerves, adi- NKCA [79]. Chronic exercise (wheel running 2 Endogenous Opiates and Exercise-Related Hypoalgesia for 5 weeks) in spontaneously hypertensive rats enhanced NKCA. The βE levels in CSF increased after the running and enhanced lymphoma cell clearance from the lungs. The deltareceptor antagonist naltrindole significantly but not completely inhibited the enhanced NKCA after 5 weeks of exercise. Neither α nor β receptor antagonists influenced the NKCA. These authors suggested that the endurance training mediated central receptor-mediated adaptations. However, if βE levels increased in the periphery via subcutaneous administration, this did not alter NKCA in vivo [79]. In contrast, NKCA after central injection of a delta opioid receptor agonist was depressed [3]. In addition, a single injection of a mu agonist into the intracerebral ventricle reduced NKCA activity. Furthermore, a single morphine injection into the periaqueductal area suppressed NKCA [173]. These findings suggest that central-mediated βE levels may act to modulate NKCA via both delta and mu receptors. Clearly more research with human models is needed, but this may be difficult as most of these actions appear to be centrally mediated. Additional βE modes of action on the immune response include mononuclear cell chemotaxis [133, 166], immunoglobulin migration [146, 166], and lymphokine production [166]. Macrophages showed migration to βE levels injected into the cerebral ventricles in rats [166]. Human neutrophils demonstrated enhanced migration to β receptors when βE was infused, and this response was blocked by prior incubation with naloxone. Analogs of opioids appear to have different responses when injected into the cerebral ventricles [146]. Some may stimulate macrophages, and others may influence neutrophils. The chemotaxis response appears to be dose-dependent [133]. High doses of βE (10−3 M) inhibited the chemotaxis response whereas low concentrations stimulated upregulation of neutrophils. Since physiological βE concentration is below the high-dose level utilized even when elevated by exercise or other stressors, it is likely that βE at these low levels provides a stimulatory effect on this aspect of the immune system. It was also noted that endogenous opioids which may 25 be elevated with exercise training induce a secondary antibody response in mice [83]. It was postulated that the opioid peptides such as βE and the enkephalins have a similar structural component to that of interleukin-2 [76]. Interleukin-2 and other interleukins are involved in the inflammatory response and are targets of βE levels and cortisol. It is highly likely that both βE levels and cortisol influence immune responses by interacting with interleukins [174]. The inhibitory response may act at a number of levels including the attenuation of production of both interleukin-1 and interleukin-6 in a dose-­ dependent manner. It appears that βE may act on a number of immune factors both centrally and peripherally and may act through both opioid and non-opioid receptors. Additionally, βE action may work through direct inhibition of cortisol. Both βE and cortisol influence immune function with βE generally enhancing immune function and cortisol acting as an immunosuppressant. The interplay of βE and cortisol in regulating immune function in response to both acute and chronic exercise requires more research to clarify their contributions. Training adaptation effects also need further study. In addition, nutritional factors (i.e., carbohydrate level) have not been adequately examined in relation to both βE and cortisol influence on immune responses with exercise. It was reported that βE increased to a similar level after cycling to exhaustion (90% VO2 max after cycling for 60 minutes at 65% VO2 max) independent of a high or a low glycemic diet or placebo prior to the exercise [72]. More studies are needed to clarify the role of diet on the βE response to exercise. ndogenous Opioids and Pain E Perception There are numerous citations that have implicated endogenous opioids and pain perception. A good number of these have suggested that endogenous opioids are involved in the processes of myocardial ischemia and or angina [74, 156]. It was reported that endorphins could modulate 26 adenosine-provoked angina pectoris-like pain in a dose-dependent manner in seven healthy subjects [156]. In contrast, met-enkephalin had no apparent effect on the pain. There may be a gender difference as angina pectoris pain induced by adenosine was attenuated by βE in males (both healthy and with coronary artery disease), but βE infusion did not modulate the pain nor did naloxone in females [144]. Increased plasma concentrations of βE were shown to alter peripheral pain threshold but did not alter angina threshold in patients with stable angina pectoris [74]. Therefore, peripheral pain may be influenced by βE, and the βE level may in part manifest some alteration in pain threshold. However, it is more likely that peripheral nerves which contain βE and/or immunocytes which release βE are involved with altering pain perception and reduction of damage [124]. Several studies have reported that exercise can modulate pain perception, and this has been attributed to endogenous opioids. Both acute and chronic exercise was reported to significantly enhance MOR expression in the hippocampal formation [27]. However, acute and chronic exercise had no significant effect on MOR expression in trained rats. Immunohistochemical techniques showed a higher number of MOR-positive cells after acute exercise compared to a control group. These authors noted that both acute and chronic exercise modulate MOR expression in the hippocampus region of rats. Higher pain thresholds for pain were reported in individuals who exercised for both finger and dental pulp stimulations [29]. Plasma βE levels increased after exercise to exhaustion as did cortisol and catecholamines, but pain threshold level changes did not correlate with plasma βE. Furthermore, naloxone failed to affect pain thresholds, despite the fact that with naloxone and exercise, βE levels increased to a greater extent. These authors suggested that the pain-related changes with exercise were not directly related to plasma βE. Janal et al. reported that after a 6.3 mile run at 85% VO2 max, hypoanalgesic effects to thermal, ischemic, and cold-­ pressor pain occurred, together with elevated mood [73]. In this study, naloxone infusion partially inhibited some of the pain and mood effects A. H. Goldfarb et al. with the exercise. This suggests that exercise can modulate pain, and it appears it is related to βE but may not be related to the plasma βE concentration. Perception of pain in trained men (N = 17) after a run (12 minute for maximal distance) with either placebo or with naloxone was examined [134]. Post-exercise βE levels increased to a similar extent for both trials, but pain level was greater with the naloxone treatment. These authors concluded that the perception of pain associated with exhaustive exercise may be related to endogenous opiates, but this had no effect on performance. Low-intensity exercise reversed muscle pain in rats, and this was blocked by naloxone [6]. Microinjections of opiates into the periaqueductal gray matter in the brain of rats attenuated pain symptoms [152]. It was found that systemic and supraspinal opiates could suppress pain in rats [106]. These studies clearly suggest that pain can be altered by opiates and that exercise can modify pain; however, the alteration in pain does not appear to be related to circulating βE. Neuropathy-induced mechanical hypersensitivity occurred in wild-type mice subjected to a chronic constriction injury of the sciatic nerve [102]. It was reported that T lymphocytes infiltrating the injury site (11% of total immune cells) released βE. Corticotropin-releasing factor (CRF) was applied at the injured nerve site and fully reversed the hypersensitivity. These authors suggested that the T lymphocytes which contain βE are crucial for not only immune function but also altered pain with peripheral nerves. It is now clear that βE is found in parts of the immune system and can act both centrally and peripherally to help modulate pain. It is unclear how these different areas in the body respond to both acute and chronic exercise, but it appears that βE are involved. Part of the modulation of pain perception is clearly related to MOR within the brain, and more research is needed to understand the effects of both acute and chronic exercise on these receptors. In addition, circulating βE may increase, but this may not always be related to pain modification, and naloxone may not always block this effect. Therefore, the 2 Endogenous Opiates and Exercise-Related Hypoalgesia peripheral-mediated βE effect on pain thresholds may not be related to the MOR in the periphery. β-Endorphin and Glucoregulation The opioid system has been implicated in the control of blood glucose concentration during rest [36, 142] and exercise [37, 71, 72]. βE and opiate receptors have been isolated from sites that are involved in glucoregulation [173]. Additionally, it has been reported that βE appears to play a role in metabolic regulation during exercise or muscle contraction [132, 137]. A bolus injection of βE followed by intravenous infusion of βE in rats raised βE levels 6–7-fold and resulted in higher plasma glucose levels at 60 and 90 minutes of exercise compared to saline infusion [37]. Lower insulin and higher glucagon levels were evident compared to saline infused rats at these times. Additionally βE exerts an effect on insulin and glucagon at rest [36, 132] in humans and animals. βE infusion without a bolus infusion of βE compared to saline infusion enhanced glucose homeostasis and exacerbated the glucagon rise in rats that were exercised [71]. This study reported that βE infusion independent of a βE bolus during exercise can attenuate blood glucose decline and increase glucagon levels in response to exercise. Additionally, βE infusion alone did not alter insulin, catecholamines, corticosterone, or FFA’s response during exercise. It appears that βE infusion alone at a level to increase circulating βE at 2.5-fold greater than normal level does not inhibit insulin; however, if the βE level increased to greater than 2.5-fold (infusion and/or increase by exercise), inhibition of insulin occurred possibly related to help maintain blood glucose. I nfluence of Acute Exercise on Enkephalins There is some evidence that exercise can increase enkephalin concentration and or opioid receptor numbers in the brain [18, 27]. These alterations in the brain have been linked to changes in mood 27 state [46], control of exercise blood pressure [7, 70, 75, 125], cardiac ischemia and angina [156], pain [154, 156], and immune function [13]. However, some of the actions of these opioid molecules may manifest themselves in other compartments such as vascular control. Research is unfolding regarding the actions of these enkephalins and enkephalin-like molecules. For example, proenkephalin peptide F which is primarily released from the adrenal gland and co-­released with epinephrine has immune-modulating functions [13, 81, 160]. Met-enkephalin level was unchanged after a Nordic ski race determined in both highly trained (N = 11, 150 km/week with greater than 3 years of experience) or recreationally trained (N = 6, 20 km/week with no competitive experience) skiers [122]. The distance covered was 75.7 km, and subjects were allowed to have water and food ad libitum. Met-enkephalin plasma concentration was determined at rest prior to a graded treadmill exercise to exhaustion and after a run of 87.2 km (5 minutes post exercise). The basal level of enkephalin was 171.7 ± 7.16 fmol/mL and increased after the treadmill exercise to 265.8 ± 9.88 fmol/mL with a further increase after the run to 378.3 ± 15.16 fmol/mL. The authors suggested that the increase in met-­ enkephalin in plasma may be related to intensity and duration of exercise [153]. The same authors compared unfit (N = 24) and fit (N = 23) subjects exposed to a graded intensity treadmill run to exhaustion (4 minute stages of at least five stages). Plasma Met-enkephalin concentration was lower for the unfit compared to the fit (126.3 ± 5.3 fmol/mL vs. 156.7 ± 6.9 fmol/mL). Both groups demonstrated increased plasma met-­ enkephalin after the exercise with the fit group showing a greater response (unfit = 180.4 ± 5.3 fmol/mL vs. fit = 278 ± 6.58 fmol/mL) [154]. In contrast, Boone et al. reported that ­met-­enkephalin was no different in trained and untrained subjects following 4 minutes of exercise at 70% VO2 max and 2 minutes at 120% VO2 max [8]. These authors noted that cryptic met-­enkephalin (activated) was elevated similarly in both groups after the 70% VO2 max and returned to baseline levels at the higher workload. 28 The response to exercise in met-enkephalin concentration in the plasma from trained and untrained subjects was reported to be similar [75]. Subjects rested for at least 15 minutes prior to collection of a resting blood sample and then performed a graded treadmill protocol to maximum, after which another blood sample was attained. There was no difference in the met-­ enkephalin concentration in plasma, red cells, cytoplasm, or ghosts when comparing pre- to post-exercise in both trained and untrained groups. However, the degradation rate was slower in the trained group compared to the untrained group independent of time (pre- and post-­ exercise). The authors suggested this may facilitate opioid responses and could provide tolerance for trained subjects. One of the early investigations in this area examined leu-enkephalin activity in plasma both before and after a competitive run [34]. Blood samples were obtained from experience runners (9 males; 5 females) before and after a 10 mile road race (2–8 minutes). Resting leu-enkephalin was 22.2 ± 13.7 pmol/mL and increased (p > 0.05) to 26.1 ± 21.5 pmol/mL, a modest increase. The leu-enkephalin change was inconsistent and variable among the runners. In conclusion, the influence of exercise on met-enkephalin is variable and appears to depend on assay method. There is inconsistency in the results, as some studies suggest enhanced levels and others no change. There is insufficient data to suggest that aerobic capacity or fitness level alters met-enkephalin level. Additionally, leu-­ enkephalin research suggests a modest increase in blood concentration with large individual variation responses with limited research. There is limited information on exercise training programs with enkephalins. Chen et al. examined acute and chronic exercise training effects on leu-enkephalin in the caudate-putamen of rat brains and compared the levels to sedentary control rats [18]. The trained rats exercised for 5 weeks on a motorized treadmill with a progressive overload in time and speed and ran 5–7 days per week. Staining of leu-enkephalin was primarily in the PVN and the caudate-putamen region (CPR). Acute exercise increased staining in the A. H. Goldfarb et al. CPR region and remained elevated in this region for up to 180 minutes post-exercise with a gradual decrease over time [18]. These results suggest that there is a central-mediated enkephalin response influencing the brain to the acute exercise in these brain regions. It also suggests that this response is transitory and reverts back to normal over time. Unfortunately, this study did not include a sedentary acute exercise group to determine whether the endurance training elicited different results than an acute exercise bout. There is also limited information with regard to the influence of exercise on proenkephalin peptide F that is typically released from the adrenal medulla and co-released with epinephrine [108]. The influence of exercise intensity and training was examined in college-aged students [100]. The trained subjects were middle-distance runners (N = 10) and were compared to untrained individuals (N = 10). The subjects exercised on a cycle ergometer for 8 minutes stages that elicited 28%, 54%, and 84% VO2 max and then exercised to VO2 max. Peptide F levels at rest were twice as high in the trained group compared to untrained but were very low (<0.1 pmol/mL). Neither group demonstrated any change in peptide F at the lowest workload, but there was a significant increase at 54% workload in the trained group. Peptide F stayed at a fairly constant concentration at the higher work intensities (~0.4 pmol/mL). In contrast, the untrained group demonstrated an increase in peptide F at 100% VO2 max that was similar to the level of the trained group. It is interesting to note that the epinephrine level for both groups showed a similar response. This suggests that peptide F level may be related to other factors than its release and epinephrine level. The effect of fitness and intensity of exercise was examined in women to see if peptide F levels might be altered differently in women [160]. Women who were endurance trained (>3 times per week, 30–45 minutes/session) were compared to inactive women. They were tested on a cycle ergometer at 60% and 80% VO2 max (15 minutes at each workload) during the early follicular phase of the menstrual cycle. Blood was collected at rest and 10 minutes into each intensity. Only the fit women demonstrated a sig- 2 Endogenous Opiates and Exercise-Related Hypoalgesia nificant increase in peptide F at the 80% intensity workload. However, this increase was modest (0.046–0.056 pmol/mL). In contrast, untrained women showed a greater epinephrine level compared to the fit women. This again suggests dissociation in the amount of epinephrine and peptide F within the circulation. The menstrual cycle effect on peptide F to maximal exercise was reported in eumenorrheic women (N = 8) [99]. There appeared to be a slight but insignificant (0.06) effect of menstrual cycle on plasma peptide F level at rest. In addition, there was no exercise main effect on plasma peptide F levels. These results suggest there may be fluctuations in peptide F levels over time as well as over the course of the menstrual cycle. This also suggests that the changes in the previous study with lower peptide F levels may be an anomaly. Clearly, more research studies with exercise on peptide F levels. Furthermore, many of the variables that might influence baseline peptide F levels should be considered. Exercise-Induced Hypoalgesia Physical activity is known to be critical for health, longevity, and high quality of life [129] and is an effective treatment as well as prevention of certain diseases [19, 77, 110, 145]. Chronic exercise has been shown to increase coordination and aerobic fitness, decrease risk of various cardiovascular diseases [112, 150], and enhance body image, self-efficacy, and emotional stability while alleviating depression and reducing anxiety and stress [54]. In addition, research has also indicated health benefits associated with acute exercise. Some forms of acute exercise influence pain perception by decreasing pain sensitivity in healthy individuals following a bout of exercise [88, 128]. This decrease in pain sensitivity, which is known as exercise-induced hypoalgesia (EIH), occurs during and after higher intensities and longer periods of aerobic exercise [66, 89]. Recent evidence suggests that signaling molecules carried in the circulation or through nerves are important for exercise-induced hypoalgesia. Jones et al. measured pressure pain thresholds in 29 subjects 5 minutes after high-intensity cycling with one arm occluded and the other with normal blood flow [78]. The investigators reported that a reduced EIH effect occurred in the occluded arm. This analgesic phenomenon is of great interest as exercise regimens are becoming the focal point of most pain management programs [141]. A review of the literature on EIH reveals that healthy individuals will demonstrate hypoalgesia following most modalities of exercise including aerobic, isometric, and dynamic resistance exercise [2, 124]. However, there are differences in the degree to which each modality alters pain perception. According to the results from a meta-­ analysis, aerobic exercise produces EIH in response to both pressure and thermal pain stimuli and seemed to be the strongest when performed at moderate-to-high intensity [124]. Isometric exercise produced the largest effect size of the modalities, and this was consistent regardless of pain stimulus and exercise intensity. There is a paucity of findings [2, 39, 90] on dynamic resistance exercise, and the effect sizes were large when pain was assessed immediately after exercise. While these studies have provided great insight into the effects of dynamic resistance exercise on EIH, there are a few key aspects of these studies that should be addressed to elucidate the effects of dynamic resistance exercise on pain perception. The first important discrepancy in research methodology is the inconsistencies in time points in which pain was assessed after exercise. Koltyn and Arbogast [90] measured pain perception at 5 and 15 minutes post-exercise, whereas Focht and Koltyn [39] assessed pain at 1 and 15 minutes time points, and Baiamonte et al. [2] utilized all three time points (1, 5, and 15 minutes post exercise). In addition, the exercise protocol implemented in each study varied in terms of sets, repetitions, intensity, and duration. Baiamonte et al. utilized 9 lifts and participants were required to perform three sets of 12 repetitions at 60% 1-RM for 45 minutes with a 1:1 work to rest ratio, while the previous two studies consisted of only four movements of three sets of 10 repetitions at 75% 1-RM for 45 minutes where the work to rest ratio was unclear but appears to be longer rest [2]. The structure of the resistance 30 A. H. Goldfarb et al. exercise protocol probably influences pain per- beta-endorphin and encephalin [55]. There is eviception and the mechanisms responsible for dence that patients with chronic low back pain EIH. In summary, all three modalities of exercise have greater activity in pain-related areas of the produced moderate-to-large effects in healthy brain, whereas there is reduced activity in analgeindividuals depending on the protocol. The EIH sic regions of the brain [101]. Recent evidence effects were transient with optimal resistance for fibromyalgia patients suggests that after ten exercise dose along with mechanisms responsi- treatments of transcranial direct current stimulable for this phenomenon still unclear. tion, pain was reduced, mood was improved, and Aerobic exercise and resistance exercise are these changes were related to circulating concenknown to elicit EIH for a brief period [2, 124]. trations of beta-endorphin [87]. Since aerobic exercise [46, 47, 48] and resistance Chronic pain disorders such as lower back exercise [56] of high enough intensity [46, 95, pain [167] and fibromyalgia [11, 114] are often 96] are known to enhance circulating endogenous treated with exercise therapy [127]. Patients with opioids, it has been speculated that EIH is due to low back pain have significant pain reduction folpain modulating substances such as beta-­ lowing treatments of aquatic exercise [151], and endorphin [88, 90, 118, 127]; however, this has unsupervised, low volume trunk exercises have not been fully supported. While the EIH findings also been shown to reduce pain in these individuhave been consistent for these exercise modali- als [57]; however, patients with long-term whipties at higher intensities in healthy participants, lash disorder do not show reductions in long-term the evidence supporting the effectiveness of exer- pain after exercise treatments [52]. cise on chronic pain patients is limited. The EIH Research has indicated that stimulation of findings in healthy individuals are more consis- afferent A-delta and C fibers via muscle contractent when compared to EIH in chronic pain popu- tions during exercise will activate spinal and lations, which produced variable outcomes with supraspinal inhibitory signaling to dampen pain small-to-large effects in individuals with regional perception [91, 158]. Both animal and human chronic pain conditions [124]. This variability studies have verified this mechanism, but findcould be explained by the exercise intensity, loca- ings have been controversial. Most studies have tion of chronic pain condition in relation to utilized administration of opioid antagonists (nalexperimental pain induction site, and severity of trexone or naloxone) prior to exercise which chronic pain condition. Interestingly, there was should bind to the mu-opioid receptors and theono evidence of EIH in patients with widespread retically prevent or reduce EIH. In both human chronic pain conditions and at times, exercise at and animal studies, the results were mixed with moderate-to-high intensity exacerbated the pain. attenuation of EIH with opioid antagonist prior to Moreover, it has been suggested that greater sen- exercise in some studies and insensitivity to opisitivity to pain in response to pressure in muscle oid antagonist in another [88]. Researchers have after static contractions in patients with fibromy- suggested that the equivocal findings are due in algia [92, 93, 129] suggests that patients with part to methodological differences which resulted fibromyalgia possibly have dysfunction of endog- in different exercise intensity, duration, and varienous analgesia during exercise compared with ations in opioid antagonist administration [91]. reduced pain sensitivity in healthy patients dur- In fact, previous research has revealed that ing exercise [94]. However, an acute exercise ses- manipulation of the exercise protocol in animal sion by women with chronic neck pain was research produce differences in EIH following shown to reduce pain intensity and sensitivity opioid antagonist administration [22]. Therefore, which was associated with greater circulating animal research has indicated that there may be beta-endorphin and cortisol concentrations [84]. multiple mechanisms (both opioid and non-­ Electroacupuncture has been used to treat opioid systems) involved in EIH [67, 91]. chronic pain and studies have reported that elec- Researchers have suggested involvement of the troacupuncture at 2 Hz will enhance release of endocannabinoid system in EIH due to the 2 Endogenous Opiates and Exercise-Related Hypoalgesia presence of CB1 receptors in pain processing areas [53, 64, 161] and evidence of increased endocannabinoid concentration after exercise [24, 41–43, 91]. A recent study by Crombie et al. [24] demonstrated an interaction between opioid system and endocannabinoid system. When participants were administered naltrexone, the endocannabinoid 2-arachidonoylglycerol (2-AG) increased significantly following exercise. Even more interesting, the endocannabinoid N-arachidonoylethanolamine (AEA) did not increase following administration of naltrexone and exercise. Therefore, increases in AEA typically observed after exercise were blocked by administration of an opioid antagonist, which suggests an interaction between the two systems. The recent work with MOR inhibition resulting in decreased voluntary wheel running in rats suggests this signaling in a dopamine-dependent manner supports complex regulation of pain at multiple levels within the brain [143]. This study noted that an overlap may at least partially explain why some individuals sense pleasure with exercise and others may not. Future research should focus on the complex interplay between the opioid and non-opioid systems on EIH rather than concentrating on each system independently. Investigation into the interaction between these two systems and probably other pathways should provide further evidence of the multiple mechanisms involved in EIH. This should provide insights into more appropriate treatments for prescribing dose and exercise intensity needed to take advantage of all the physical and mental benefits that exercise has to offer besides just EIH. -Endorphin and Pain in Clinical β Populations There are a number of studies that investigated the effects of exercise on βE in different clinical populations affected by pain. Circulating concentrations of several neuropeptides, steroid hormones and metabolites were assessed after exercise to determine if women with chronic neck/shoulder pain responded differently than 31 healthy women [84]. The investigators used microdialysis to analyze substance P, βE, cortisol, glutamate, lactate, and pyruvate before and after an exercise training regimen. They also assessed pain intensity and pain threshold. Before the training regimen, women with neck/shoulder pain had higher circulating levels of glutamate and βE and lower cortisol concentration than healthy women. Following exercise training program, women with shoulder/neck pain had less circulating substance P (and possibly glutamate) and greater circulating concentrations on βE and cortisol as well as reduced pain intensity and higher pain pressure thresholds. The researchers suggested that exercise training could alter pain intensity and sensitivity as well as peripheral substances related to pain. This study provides more suggestive effects of opioid-mediated pain modification following exercise training. In a study conducted on coronary artery bypass graft surgery patients, transcutaneous electrical nerve stimulation (TENS) or sham TENS was applied over the posterior cervical region (C7-T4) to access the stellate ganglion region, 5 days after surgery [20]. The treatment was conducted four times per day for 30 minutes per session. Patients who had TENS treatment reported less postoperative pain and had less opiate requirements with higher circulating βE. They also had greater limb blood flow during a sympathetic stimulation (cold pressor) procedure. Thus, TENS, which elicits muscle contractions, appears to increase circulating βE which could lead to pain reduction. dvanced Techniques to Investigate A β-Endorphin and Pain Thermal heat pain challenges were employed before and after running and walking trials to determine the effects of exercise intensity on pain [155]. The pattern of pain-related activity in response to heat/pain treatment using fMRI analysis was compared. The medial and lateral pain systems and periaqueductal gray (PAG) were key areas of the descending antinociceptive pathway that were evaluated. Running reduced affective A. H. Goldfarb et al. 32 pain ratings whereas walking did not. fMRI revealed that there was a reduction in pain activation in the PAG with decreases after running but pain activation was elevated after walking. For the pregenual anterior cingulate cortex and middle insular cortex there were similar trends of activation for running vs. walking. Importantly, the authors concluded that increased circulating βE levels that were noted with running, but not walking suggested involvement of the opioidergic system. Another study which utilized fMRI examined pain modulation in athletes both before and after running or walking [147]. They examined both PAG and pain ratings and noted enhanced antinociceptive mechanisms were attenuated by running (23 km, HR = 148) but not walking (10 km, HR = 84). Elevated plasma beta-­ endorphins were reported only after running. These results support previous studies that indicated sufficient exercise intensity and duration are needed to influence blood beta-endorphin levels. A recent fMRI study reported that resistance exercise training (twice per week for 15 weeks) in fibromyalgia patients did not significantly alter distraction-induced analgesia nor influence brain activity [115]. This group previously reported the influence of resistance exercise training in a larger cohort of subjects [105]. responsible for pain reduction following bouts of exercise. The most commonly proposed mechanism includes activation of the endogenous opioid system, in particular the release of βE with the CNS [66] and from muscle contractions during intense exercise stimulating pain receptors in skeletal muscle which can stimulate the endogenous opioid system [163]. However, previous research on humans and animals has been unclear regarding the involvement of the endogenous opioid system in EIH after opioid antagonist administration [91]. In animal studies, the opioid antagonists attenuated the hypoalgesic effect of exercise whereas human studies have produced conflicting results [91]. Therefore, further investigation into the role of endorphins in EIH is warranted to address the inconsistent findings in the literature. Recent Pain Models In a recent review, it was determined that increased pain threshold following exercise was attributed to release of endogenous opioids [127]. More specifically, EIH was demonstrated in healthy participants due to activation of μ-opioid receptors both peripherally and centrally. However, evidence of EIH in individuals with chronic pain has been equivocal. Research has Role of Enkephalins indicated that exercise of many modalities can decrease pain symptoms, resulting in improved It is possible that enkephalin peptides play a role daily function for individuals who suffer from in altering pain sensation. Exercise- or ischemia-­ chronic pain [17, 30, 51, 58, 113, 116, 147]. In induced enkephalin release from selected tissues contrast, exercise may not produce pain facilitawas examined in rat, mouse, pig, and human tis- tion in certain chronic pain groups [127]. For sues [26]. Using real-time PCR, Western blot example, patients with fibromyalgia, whiplash, analysis, ELISA, and immunofluorescence and chronic fatigue all demonstrate pain sensitivmicroscopy, they reported extensive expression ity following exercise [52, 104, 119, 168, 169]. of preproenkephalin mRNA as well as enkepha- Nijs et al. suggested that these patients had “dyslin precursor protein proenkephalin. Isolated functional endogenous analgesia” in response to ex vivo tissue that were analyzed revealed that exercise resulting in abnormalities in the central skeletal muscle, heart muscle, and intestinal tis- pain modulation system, which includes βE sue released enkephalins. The investigators con- [127]. While this topic has been extensively cluded that non-neuronal tissues could aid in investigated over the last 20 years, research has inducing local and systemic enkephalin effects. mainly focused on the hypoalgesic effects of From a physiological standpoint, most exercise following a single episode of exercise as researchers fail to agree on the exact mechanism(s) a result of increases in endogenous opioids [89]. 2 Endogenous Opiates and Exercise-Related Hypoalgesia Current research should focus on the effects of repeated exercise on chronic pain and attempt to discover the mechanisms responsible. It has been hypothesized that regular aerobic exercise leads to sustained reversal of neuropathic pain by activating endogenous opioid-mediated pain modulatory systems [157]. Following nerve ligation, rats displayed thermal and mechanical sensitivities that were attenuated within 3 weeks of exercise training [155]. However, hyperalgesia returned 5 days after cessation of exercise. These authors provided evidence of βE and met-­ enkephalin involvement and injected naltrexone into the intracerebroventricular region which reversed EIH. Recent studies have indicated increased βE and met-enkephalin in the medulla and periaqueductal gray area, regions of the brain that are also involved in the descending pain pathway. Conclusion Exercise of sufficient intensity and duration can induce transient pain modification, but more evidence is needed to substantiate the role of agents that are involved in triggering the mechanisms of action in EIH. βE can bind to various opioid receptors within the CNS and can modify pain. However, it is unclear if these actions are solely dependent to induce the EIH and if these actions reside exclusively within the CNS or are also triggered by agents outside the CNS such as signals arising from exercising muscles to help alleviate pain. In addition, not all individuals may respond in a similar manner, thus the proposed mechanisms explaining why EIH may work in some and not others, needs further clarification. Summary In conclusion, exercise of sufficient intensity and duration may influence the endogenous opioids, but what is measured in the circulation does not necessarily reflect what occurs within the brain. Numerous factors such as sex, menstrual cycle, 33 diet, plasma volume, carbohydrate level, and inflammation can influence the endogenous opioids. Furthermore, immune function and neural control can clearly alter endogenous opioid activity. Finally, a greater understanding of the influence of exercise on the endogenous opioid effects in the brain needs to be established. References 1. Angelopoulos TJ. Beta-endorphin immunoreactivity during high-intensity exercise with and without opiate blockade. Eur J Appl Physiol. 2001;86(1):92–6. 2. Baiamonte BA, Kraemer RR, Chabreck CN, et al. Exercise-induced hypoalgesia: Pain tolerance, preference and tolerance for exercise intensity, and physiological correlates following dynamic circuit resistance exercise. J Sports Sci. 2017;35(18):1–7. 3. Band LC, Pert A, Willams W, et al. Central μ-opioid receptors mediate suppression of natural killer activity in vivo. Prog Neuroendocr. 1992;5:95–101. 4. Bardoni R, Tawfik VL, Wang D, et al. Delta opioid receptors presynaptically regulate cutaneous mechanosensory neuron input to the spinal cord dorsal horn. Neuron. 2014;81:1312–27. 5. Basbaum AI, Bautista DM, Scherrer G, Julius D. Cellular and molecular mechanisms of pain. Cell. 2009;139:267–84. 6. Bement MK, Sluka KA. Low-intensity exercise reverses chronic muscle pain in the rat in a naloxone-­ dependent manner. Arch Phys Med Rehabil. 2005;86:1736–40. 7. Boone JB Jr, Corry JM. Proenkephalin gene expression in the brainstem regulates post-exercise hypotension. Brain Res Mol Brain Res. 1996;42(1):31–8. 8. Boone JB Jr, Sherraden T, Pierzchala K, et al. Plasma Met-enkephalin and catecholamine responses to intense exercise in humans. J Appl Physiol. 1992;73(1):388–92. 9. Bradbury AF, Smyth DG, Snell CR, et al. C fragment of lipotropin has a high affinity for brain opiate receptors. Nature. 1976;260:793–5. 10. Brellenthin AG, Crombie KM, Hillard CJ, Koltyn KF. Endocannabinoid and mood responses to exercise in adults with varying activity levels. Med Sci Sports Exerc. 2017;49(8):1688–96. 11. Brosseau L, Wells GA, Tugwell P, et al. Ottawa Panel evidence-based clinical practice guidelines for aerobic fitness exercises in the management of fibromyalgia: part 1. Phys Ther. 2008;88:857–71. 12. Bullen BA, Skrinar GS, Beitins IZ, et al. Endurance training effects on plasma hormonal responsiveness and sex hormone excretion. J Appl Physiol. 1984;56(6):1453–63. 13. Bush JA, Mastro AM, Kraemer WJ. Proenkephalin peptide F immunoreactivity in different circulatory A. H. Goldfarb et al. 34 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. biocompartments after exercise. Peptides. 2006;27(6):1498–506. Carr DB, Ballantyene JC. Denorphins and analgesia. Compr Ther. 1987;13(12):7–13. Carr DB, Bullen BA, Skrinar GS, et al. Physical conditioning facilitates the exercise-induced secretion of beta-endorphin and beta-lipotropin in women. N Engl J Med. 1981;305:560–3. Chan HCS, McCarthy D, Li J, et al. Designing safer analgesics via μ-opioid receptor pathways. Trends Pharmacol Sci. 2017;38:1016–37. Chatzitheodorou D, Kabitsis C, Malliou P, Mougios V. A pilot study of the effects of high-intensity aerobic exercise versus passive interventions on pain, disability, psychological strain, and serum cortisol concentrations in people with chronic low back pain. Phys Ther. 2007;87(3):304–12. Chen JX, Zhao X, Yue GX, et al. Influence of acute and chronic treadmill exercise on rat plasma lactate and brain NPY, L-ENK, DYN A1-13. Cell Mol Neurobiol. 2007;27(1):1–10. Clark JE. Diet, exercise or diet with exercise: comparing the effectiveness of treatment options for weight-loss and changes in fitness for adults (18-­ 65 years old) who are overfat, or obese; systematic review and meta-analysis. J Diabetes Metab Disord. 2015;14:31. Cipriano G, Neder JA, Umpierre D, et al. Sympathetic ganglion transcutaneous electrical nerve stimulation after coronary artery bypass graft surgery improves femoral blood flow and exercise tolerance. J Appl Physiol (1985). 2014;117(6):633–8. Colt EW, Wardlaw SL, Frantz AG. The effect of running on plasma beta-endorphin. Life Sci. 1981;28:1637–40. Cook DB, Koltyn KF. Pain and exercise. Int J Sport Psychol. 2000;31:256–77. Corder G, Tawfik VL, Wang D, et al. Loss of μ opioid receptor signaling in nociceptors, but not microglia, abrogates morphine tolerance without disrupting analgesia. Nat Med. 2017;23:164–73. Crombie KS, Brellenthin AG, Hillard CJ, Koltyn KF. Endocannabinoid and opioid system interactions in exercise-induced hypoalgesia. Pain Med. 2017;19(1):118–23. https://doi.org/10.1093/pm/ pnx058. Dearman J, Francis KT. Plasma levels of catecholamines, cortisol, and beta-endorphins in male athletes after running 26.2, 6, and 2 miles. J Sports Med Phys Fitness. 1983;23:30–8. Denning GM, Ackermann LW, Barna TJ, et al. Proenkephalin expression and enkephalin release are widely observed in non-neuronal tissues. Peptides. 2008;29(1):83–92. de Oliveira MS, da Silva Fernandes MJ, Scorza FA. Acute and chronic exercise modulates the expression of MOR opioid receptors in the hippocampal formation of rats. Brain Res Bull. 2010;83(5):278–83. 28. Donevan RH, Andrew GM. Plasma beta-endorphin immunoreactivity during graded cycle ergometry. Med Sci Sports Exerc. 1987;19(3):229–33. 29. Droste C, Greenlee MW, Schreck M, et al. Experimental pain thresholds and plasma beta-­ endorphin levels during exercise. Med Sci Sports Exerc. 1991;23(3):334–42. 30. Ellingson LD, Koltyn KF, Kim JS, Cook DB. Does exercise induce hypoalgesia through conditioned pain modulation? Psychophysiology. 2014;51(3):267–76. 31. Elliot DL, Goldberg L, Watts WJ, et al. Resistance exercise and plasma beta-endorphin/beta-­lipotrophin immunoreactivity. Life Sci. 1984;34(6):515–8. 32. Engfred K, Kjaer M, Secher NH, et al. Hypoxia and training-induced adaptation of hormonal responses to exercise in humans. Eur J Appl Physiol Occup Physiol. 1994;68(4):303–9. 33. Farrell PA, Gates WK, Maksud MG, et al. Increases in plasma beta-endorphin/beta-lipotropin immunoreactivity after treadmill running in humans. J Appl Physiol. 1982;52(5):1245–9. 34. Farrell PA, Gates WK, Morgan WP, Pert CB. Plasma leucine enkephalin-like radioreceptor activity and tension-anxiety before and after competitive running. In: Knuttgen HG, Vogel JA, Poortmans J, editors. Biochemistry of exercise, vol. 13. Champaign: Human Kinetics; 1983. p. 637–44. 35. Farrell PA, Kjaer M, Bach FW, et al. Beta-endorphin and adrenocorticotropin response to supramaximal treadmill exercise in trained and untrained males. Acta Physiol Scand. 1987;130(4):619–25. 36. Fatouros J, Goldfarb AH. Low carbohydrate diet induces changes in central and peripheral beta-­ endorphins. Nutr Res. 1995;15(11):1683–94. 37. Fatouros J, Goldfarb AH, Jamurtas AZ, et al. Beta-­ endorphin infusion alters pancreatic endocrines and plasma glucose during exercise in rats. Eur J Appl Physiol. 1997;76:203–8. 38. Ferreira MD, Menescal-de-Oliveira L. Opioidergic, GABAergic and serotonergic neurotransmission in the dorsal raphe nucleus modulates tonic immobility in guinea pigs. Physiol Behav. 2012;106(2):109–16. 39. Focht BC, Koltyn K. F. Alterations in pain perception after resistance exercise performed in the morning and evening. J Strength Cond Res. 2009;23(3):891–7. 40. Fry AC, Bonner E, Lewis DL, et al. The effects of gamma-oryzanol supplementation during resistance exercise training. Int J Sport Nutr. 1997;7(4):318–29. 41. Galdino G, Romero TR, Silva JF, et al. The endocannabinoid system mediates aerobic exercise-induced antinociception in rats. Neuropharmacology. 2014;77:313–24. 42. Galdino GS, Romero T, Silva JF, et al. Acute resistance exercise induces antinociception by activation of the endocannabinoid system in rats. Anesth Analg. 2014;119(3):702–15. 43. Gannon GA, Rhind SG, Suzui M, et al. Beta-­ endorphin and natural killer cell cytolytic activity 2 Endogenous Opiates and Exercise-Related Hypoalgesia 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. during prolonged exercise is there a connection? Am J Phys. 1998;275(6 Pt 2):1725–34. Gilman SC, Schwartz JM, Milner RJ, et al. Beta-endorphin enhances lymphocyte proliferative responses. Proc Natl Acad Sci U S A. 1982;79(13):4226–30. Goldfarb AH. Effect of gender and menstrual cycle on Beta-endorphin response to activity. Am J Med Sports. 2001;3:363–6. Goldfarb AH, Hatfield BD, Armstrong D, et al. Plasma beta-endorphin concentration: response to intensity and duration of exercise. Med Sci Sports Exerc. 1990;22:241–4. Goldfarb AH, Hatfield BD, Potts J, et al. Beta-­ endorphin at the same relative exercise intensity: training effects. Int J Sports Med. 1991;12(3):264–8. Goldfarb AH, Hatfield BD, Sforzo GA, et al. Serum beta-endorphins levels during a graded exercise test to exhaustion. Med Sci Sports Exerc. 1987;19(2):78–82. Goldfarb AH, Jamurtas AZ, Kamimori G, et al. Gender and menstrual cycle effects on circulating beta-endorphin in response to exercise. Med Sci Sports Exerc. 1998;30(12):1672–6. Goldstein A, Tachibana S, Lowney LI, et al. Dynorphin-(1–13), an extraordinarily potent opioid peptide. Proc Natl Acad Sci U S A. 1979;76(12):6666–70. Gowans SE, Dehueck A, Voss S, et al. Six-month and one-year follow up of 23 weeks of aerobic exercise for individuals with fibromyalgia. Arthritis Rheum. 2004;51(6):890–8. Griffin A, Leaver A, Moloney N. General exercise does not improve long-term pain and disability in individuals with whiplash-associated disorders: a systematic review. J Orthop Sports Phys Ther. 2017;47(7):472–80. Guindon J, Beaulieu P. The role of the endogenous cannabinoid system in peripheral analgesia. Curr Mol Pharmacol. 2009;2(1):134–9. Gurevich M, Kohn PM, Davis C. Exercise-induced analgesia and the role of reactivity in pain sensitivity. J Sports Sci. 1994;6:549–59. Han JS. Acupuncture and endorphins. Neurosci Lett. 2004;361(1–3):258–61. Harber VJ, Sutton JR, MacDougall JD, et al. Plasma concentrations of beta-endorphin in trained eumenorrheic and amenorrheic women. Fertil Steril. 1997;67(4):648–53. Haufe S, Wiechmann K, Stein L, et al. Low-dose, non-supervised, health insurance initiated exercise for the treatment and prevention of chronic low back pain in employees. Results from a randomized controlled trial. PLoS One. 2017;12(6):e0178585. Hayden JA, Van Tulder MW, Tomlinson G. Systematic review: strategies for using exercise therapy to improve outcomes in chronic low back pain. Ann Intern Med. 2005;142(9):776–85. 35 59. Hebbes C. Non-opioid analgesics. Anaesth Intensive Care Med. 2016;17(9):469–72. 60. Hedner T, Cassuto J. Opioids and opioid receptors in peripheral tissues. Scand J Gastroenterol Suppl. 1987;130:27–46. 61. Heitkamp HC, Huber W, Scheib K. Beta-endorphin and adrenocorticotrophin after incremental exercise and marathon running—female responses. Eur J Appl Physiol Occup Physiol. 1996;72(5–6):417–24. 62. Heitkamp HC, Schmid K, Scheib K. Beta-endorphin and adrenocorticotropic hormone production during marathon and incremental exercise. Eur J Appl Physiol Occup Physiol. 1993;66(3):269–74. 63. Hemmick LM, Bidlack JM. Beta-endorphin stimulates rat T lymphocyte proliferation. J Neuroimmunol. 1990;29(1–3):239–348. 64. Herkenham M, Lynn AB, Johnson MR, et al. Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci. 1991;11:563–83. 65. Hoffman MD, Shepanski MA, Ruble SB, et al. Intensity and duration threshold for aerobic exercise-­ induced analgesia to pressure pain. Arch Phys Med Rehabil. 2004;85:1183–7. 66. Hoffman P, Terenius L, Thoren P. Cerebrospinal fluid immunoreactive beta-endorphin concentration is increased by voluntary exercise in the spontaneously hypertensive rat. Regul Pept. 1990;28:233–9. 67. Hohmann AG, Suplita RL. Endocannabinoid mechanisms of pain modulation. AAPSJ. 2006;9:E693–708. 68. Howlett TA, Tomlin S, Ngahfoong L, et al. Release of beta endorphin and met-enkephalin during exercise in normal women: response to training. Br Med J (Clin Res Ed). 1984;288(6435):1950–2. 69. Hughes J, Smith TW, Kosterlitz HW, et al. Identification of two related pentapeptides from the brain with potent opiate agonist activity. Nature. 1975;258:577–9. 70. Ishide T, Mancini M, Maher TJ, et al. Rostral ventrolateral medulla opioid receptor activation modulates glutamate release and attenuates the exercise pressor reflex. Brain Res. 2000;865(2):177–85. 71. Jamurtas AZ, Goldfarb AH, Chung SC, et al. Beta-endorphin infusion during exercise in rats: blood metabolic effects. Med Sci Sports Exerc. 2000;32(9):1570–5. 72. Jamurtas AZ, Tofas T, Fatouros I, et al. The effects of low and high glycemic index foods on exercise performance and beta-endorphin responses. J Int Soc Sports Nutr. 2011;8(1):15. 73. Janal MN, Colt EW, Clark WC, et al. Pain sensitivity, mood and plasma endocrine levels in man following long-distance running: effects of naloxone. Pain. 1984;19(1):13–25. 74. Jarmukli NF, Ahn J, Iranmanesh A, et al. Effect of raised plasma beta endorphin concentrations on peripheral pain and angina thresholds in patients with stable angina. Heart. 1999;82(2):204–9. A. H. Goldfarb et al. 36 75. Jaskowski MA, Jackson AS, Raven PB, et al. Enkephalin metabolism: effect of acute exercise stress and cardiovascular fitness. Med Sci Sports Exerc. 1989;21(2):154–60. 76. Jiang CL, Xu D, Lu CL, et al. Interleukin-2: structural and biological relatedness to opioid peptides. Neuroimmunomodulation. 2000;8(1):20–4. 77. Johnson EJ, Dieter BP, Marsh SA. Evidence for distinct effects of exercise in different cardiac hypertrophic disorders. Life Sci. 2015;123:100–6. 78. Jones MD, Taylor JL, Barry BK. Occlusion of blood flow attenuates exercise-induced hypoalgesia in the occluded limb of healthy adults. J Appl Physiol. 2017;122:1284–91. 79. Jonsdottir IH, Hellstrand K, Thorén P, et al. Enhancement of natural immunity seen after voluntary exercise in rats. Role of central opioid receptors. Life Sci. 2000;66(13):1231–9. 80. Kahn S, Anthony A, Hughes S, et al. Beta-endorphin decreases fatigue and increases glucose uptake independently in normal and dystrophic mice. Muscle Nerve. 2005;31:481–6. 81. Kamphuis S, Eriksson F, Kavelaars A, et al. Role of endogenous pro-enkephalin A-derived peptides in human T cell proliferation and monocyte IL-6 production. J Neuroimmunol. 1998;84:53–60. 82. Kandasamy R, Price TJ. The pharmacology of nociceptor priming. Handb Exp Pharmacol. 2015;227:15–37. 83. Kapasi ZF, Catlin PA, Beck J, et al. The role of endogenous opioids in moderate exercise training-­ induced enhancement of the secondary antibody response in mice. Phys Ther. 2001;81(11):1801–9. 84. Karlsson L, Gerdle B, Ghafouri B, et al. Intramuscular pain modulatory substances before and after exercise in women with chronic neck pain. Eur J Pain. 2015;19(8):1075–85. 85. Kay N, Allen J, Morley JE. Endorphins stimulate normal human peripheral blood lymphocyte natural killer activity. Life Sci. 1984;35(1):53–9. 86. Khachaturian H, Lewis ME, Schafer MK-H, et al. Anatomy of the CNS opioid systems. Trends Neurosci. 1985;8:111–9. 87. Khedr EM, Omran EAH, Ismail NM, et al. Effects of transcranial direct current stimulation on pain, mood, and serum endorphin level in the treatment of fibromyalgia: a double blinded randomized clinical trial. Brain Stimul. 2017;10(5):893–901. 88. Koltyn KF. Analgesia following exercise. A review. Sports Med. 2000;29(2):85–98. 89. Koltyn KF. Exercise-induced hypoalgesia and intensity of exercise. Sports Med. 2002;32(8):477–87. 90. Koltyn KF, Arbogast RW. Perception of pain after resistance exercise. Br J Sports Med. 1998;32:1. 91. Koltyn KF, Brellenthin AG, Cook DB, et al. Mechanisms of exercise-induced hypoalgesia. J Pain. 2014;15(12):1294–304. 92. Kosek E, Ekhom J, Hansson P. Increased pressure pain sensibility in fibromyalgia patients is located 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. deep to the skin but not restricted to muscle tissue. Pain. 1995;63(3):335–9. Kosek E, Ekhom J, Hansson P. Modulation of pressure pain thresholds during and following isometric contraction in patients with fibromyalgia and in healthy controls. Pain. 1996;64(3):415–23. Kosek E, Lundberg L. Segmental and plurisegmental modulation of pressure thresholds during static muscle contractions in healthy individuals. Eur J Pain. 2003;7(3):251–8. Kraemer RR, Acevedo EO, Dzewaltowski D, et al. Effects of low-volume resistive exercise on beta-­ endorphin and cortisol concentrations. Int J Sports Med. 1996;17(1):12–6. Kraemer RR, Blair S, Kraemer GR, Castracane VD. Effects of treadmill running on plasma beta-­ endorphin, corticotropin, and cortisol levels in male and female 10K runners. Eur J Appl Physiol Occup Physiol. 1989;58(88):45–51. Kraemer WJ, Dziados JE, Marchitelli LJ, et al. Effects of different heavy-resistance exercise protocols on plasma beta-endorphin concentrations. J Appl Physiol. 1993;74(1):450–9. Kraemer WJ, Fry AC, Warren BJ, et al. Acute hormonal responses in elite junior weightlifters. Int J Sports Med. 1992;13(2):103–9. Kraemer WJ, Kim SK, Bush JA, et al. Influence of the menstrual cycle on proenkephalin peptide F responses to maximal cycle exercise. Eur J Appl Physiol. 2006;96(5):581–6. Kraemer WJ, Nobles B, Culver B, et al. Changes in plasma proenkephalin peptide F and catecholamine levels during graded exercise in men. Proc Natl Acad Sci U S A. 1985;82:6349–51. Kregel J, Meeus M, Malfiet A, et al. Structural and functional brain abnormalities in chronic low back pain: a systematic review. Semin Arthritis Rheum. 2015;45(2):229–37. Labuz D, Schreiter A, Schmidt Y, et al. T lymphocytes containing β-endorphin ameliorate mechanical hypersensitivity following nerve injury. Brain Behav Immun. 2010;24(7):1045–53. Langenfeld ME, Hart LS, Kao PC. Plasma β-endorphin responses to one-hour bicycling and running at 60% VO2max. Med Sci Sports Exerc. 1987;19:83–6. Lannersten L, Kosek E. Dysfunction of endogenous pain inhibition during exercise with painful muscles in patients with shoulder myalgia and fibromyalgia. Pain. 2010;151(1):77–86. Larsson A, Palstam A, Lofgren M, et al. Resistance exercise improves muscle strength, health status and pain intensity in fibromyalgia – a randomized controlled trial. Arthritis Res Ther. 2015;17:161. https:// doi.org/10.1186/s13075-015-0679-1. Lee YW, Chaplan SR, Yaksh T. Systemic and supraspinal, but not spinal, opiates suppress allodynia in a rat neuropathic pain model. Neruosci Lett. 1995;199:111–4. 2 Endogenous Opiates and Exercise-Related Hypoalgesia 107. Li CH, Chung D. Isolation and structure of an 122. untriakontapeptide with opiate activity from camel pituitary glands. Proc Natl Acad Sci U S A. 1976;73(4):1145–8. 108. Livett BG, Dean DM, Whelan LG, et al. Co-release of enkephalin and catecholamines from cultured 123. adrenal chromaffin cells. Nature. 1981;289:317–9. 109. Lobstein DD, Ismail AH. Decreases in resting plasma beta-endorphin/-lipotropin after endurance 124. training. Med Sci Sports Exerc. 1989;21(2):161–6. 110. Lukács A, Barkai L. Effect of aerobic and anaerobic exercises on glycemic control in type 1 diabetic 125. youths. World J Diabetes. 2015;6(3):534–42. 111. Luger A, Deuster PA, Kyle SB, et al. Acute hypothalamic-­ pituitary-adrenal responses to the stress of treadmill exercise physiologic adaptations to physical training. N Engl J Med. 1987;316:1309–15. 112. Lyu X, Li S, Peng S, et al. Intensive walking exer- 126. cise for lower extremity peripheral arterial disease: A systematic review and meta-­ analysis. J Diabetes. 2015;8(3):363–77. https://doi. 127. org/10.1111/1753-0407.12304. 113. Malmros B, Mortensen L, Jensen MB, Charles P. Positive effects of physiotherapy on chronic pain and performance in osteoporosis. Osteoporosis Int. 1998;8(3):215–21. 114. Mannerkorpi K, Henriksson C. Nonpharmacological 128. treatment of chronic widespread musculoskeletal pain. Best Pract Res Clin Rheumatol. 2007;21:513–34. 115. Martinsen S, Flodin P, Berrebi J, et al. The role of 129. long-term physical exercise on performance and brain activation during the Stroop colour word task in fibromyalgia patients. Clin Physiol Funct Imaging. 2017;38(3):508–16. https://doi.org/10.1111/ 130. cpf.12449. 116. McCain GA, Bell DA, Mai FM, Halliday PD. A controlled study of the effects of a supervised cardiovascular fitness training program on the manifestations of primary fibromyalgia. Arthritis Rheum. 131. 1988;31(9):1135–41. 117. McGowan RW, Pierce EF, Eastman N, et al. Beta-­ endorphins and mood states during resistance exer132. cise. Percept Motor Skills. 1993;76(2):376–8. 118. McMurray RG, Forsythe WA, Mar MH, et al. Exercise intensity-related responses of beta-­ endorphin and catecholamines. Med Sci Sports 133. Exerc. 1987;6:570–4. 119. Meeus M, Roussel NA, Truijen S, Nijs J. Reduced pressure pain thresholds in response to exercise in chronic fatigue syndrome but not in chronic low back pain: an experimental study. J Rehabil Med. 134. 2010;42(9):884–90. 120. Metzger JM, Stein EA. Beta-endorphin and sprint training. Life Sci. 1984;34(16):1541–7. 121. Millar DB, Hough CJ, Mazorow DL, et al. Beta-­ endorphin’s modulation of lymphocyte proliferation 135. is dose, donor, and time dependent. Brain Behav Immun. 1990;4(3):232–42. 37 Mougin C, Baulay A, Henriet MT, et al. Assessment of plasma opioid peptides, beta-endorphin and met-enkephalin, at the end of an international Nordic ski race. Eur J Appl Physiol Occup Physiol. 1987;56(3):281–6. Mousa SA, Zhang Q, Sitte N, et al. β-Endorphin-­ containing memory-cells and μ-opioid receptors undergo transport to peripheral inflamed tissue. J Neuroimmunol. 2001;15(1–2):71–8. Naugle KM, Fillingim RB, Riley JL. A meta-­analytic review of the hypoalgesic effects of exercise. J Pain. 2012;13(12):1139–50. Nauli SM, Maher TJ, Pearce WJ, et al. Effects of opioid receptor activation on cardiovascular responses and extracellular monoamines within the rostral ventrolateral medulla during static contraction of skeletal muscle. Neurosci Res. 2001;41(4):373–83. Navolotskaya EV, Malkova NV, Zargarova TA, et al. Synthetic beta-endorphin-like peptide immunorphin binds to non-opioid receptors for beta-endorphin on T lymphocytes. Peptides. 2001;22(12):2009–13. Nijs J, Kosek E, Van Oosterwijck J, Meeus M. Dysfunctional endogenous analgesia during exercise in patients with chronic pain: to exercise or not to exercise? Pain Physician. 2012;3S:ES205–13. O’Connor PJ, Cook DB. Exercise and pain: the neurobiology, measurement, and laboratory study of pain in relation to exercise in humans. Exerc Sport Sci Rev. 1999;27:119–66. O’Donovan G, Blazevich A, Boreham C, et al. The ABC of physical activity for health: a consensus statement from the British association of sport and exercise sciences. J Sports Sci. 2010;28(6):573–91. Oleshansky MA, Zoltick JM, Herman RH, et al. The influence of fitness on neuroendocrine responses to exhaustive treadmill exercise. Eur J Appl Physiol Occup Physiol. 1990;59(6):405–10. Owen DL, Morley JS, Ensor DM, et al. The C-terminal tetrapeptide of beta-endorphin (MPF) enhances lymphocyte proliferative responses. Neuropeptides. 1998;32(2):131–9. Paolisso G, Giugliano D, Scheen A, et al. Primary role of glucagon release in the effect of β-endorphin on glucose homeostasis in normal man. Acta Endocrinol. 1987;115:161–9. Pasnik J, Tchorzewski H, Baj Z, et al. Priming effect of met-enkephalin and β-endorphin on chemiluminescence, chemotaxis and CD11b molecule expression on human neutrophils in vitro. Immunol Lett. 1999;67(2):77–83. Paulev PE, Thorbøll JE, Nielsen U, et al. Opioid involvement in the perception of pain due to endurance exercise in trained man. Jpn J Physiol. 1989;39(1):67–74. Pert CB, Snyder SH. Properties of opiate-receptor binding in rat brain. Proc Natl Acad Sci U S A. 1973;70(8):2243–7. 38 136. Pierce EF, Eastman NW, McGowan RW, et al. Resistance exercise decreases beta-endorphin immunoreactivity. Br J Sports Med. 1994;28(3):164–6. 137. Pierce EF, Eastman NW, Tripathi HT, et al. Plasma beta-endorphin immunoreactivity: response to resistance exercise. J Sports Sci. 1993;11(6):499–502. 138. Radosevich PM, Nash JA, Lacy DB, et al. Effects of low- and high-intensity exercise on plasma and cerebrospinal fluid levels of ir-beta-endorphin, ACTH, cortisol, norepinephrine and glucose in the conscious dog. Brain Res. 1989;498(1):89–98. 139. Rahkila P, Hakala E, Alén M, et al. Beta-endorphin and corticotropin release is dependent on a threshold intensity of running exercise in male endurance athletes. Life Sci. 1988;43(6):551–8. 140. Rahkila P, Hakala E, Salminen K, et al. Response of plasma endorphins to running exercises in male and female endurance athletes. Med Sci Sports Exerc. 1987;19(5):451–5. 141. Raithel KS. Chronic pain and exercise therapy. Phys Sportsmed. 1989;17(3):203–5. 142. Reid RL, Yen SS. Beta-endorphin stimulates the secretion of insulin and glucagon in humans. J Clin Endocrinol Metab. 1981;52(3):592–4. 143. Ruegsegger GN, Brown JD, Kovarik MC, et al. Mu opioid receptor inhibition decreases voluntary wheel running in a dopamine-dependent manner in rats bred for high voluntary running. Neuroscience. 2016;339:525–37. 144. Sadigh B, Berglund M, Fillingim RB, et al. Beta-­ endorphin modulates adenosine provoked chest pain in men, but not in women-a comparison between patients with ischemic heart disease and healthy volunteers. Clin J Pain. 2007;23(9):750–5. 145. Sahni S, Capozzi B, Iftikhar A, et al. Pulmonary rehabilitation and exercise in pulmonary arterial hypertension: an underutilized intervention. J Exerc Rehabil. 2015;11(2):74–9. 146. Saland LC, Van Epps DE, Maez D, et al. Acute infusion of chemotactic or enkephalin-analog peptides into rat cerebral ventricles: scanning and transmission electron microscopy of leukocyte immigration in vivo. J Neuroimmunol. 1988;18(3):197–206. 147. Scheef L, Jankowski J, Daamen M, et al. An fMRI study on the acute effects of exercise in pain processing in trained athletes. Pain. 2012;153(8):1702–14. 148. Schwarz L, Kindermann W. Beta-endorphin, adrenocorticotropic hormone, cortisol and catecholamines during aerobic and anaerobic exercise. Eur J Appl Physiol Occup Physiol. 1990;61(3–4):165–71. 149. Sforzo GA, Seeger TF, Pert CB, et al. In vivo opioid receptor occupation in the rat brain following exercise. Med Sci Sports Exerc. 1986;18(4):380–4. 150. Shephard RJ. Exercise and malignancy. Sports Med. 1986;3:235–41. 151. Shi Z, Zhou H, Lu L, et al. Aquatic exercises in the treatment of low back pain: a systematic review of the literature and meta-analysis of eight studies. Am J Phys Med Rehabil. 2018;97(2):116–22. A. H. Goldfarb et al. 152. Sohn JH, Lee BH, Park SH, et al. Microinjection of opiates into the periaqueductal gray matter attenuates neuropathic pain symptoms in rats. Neuroreport. 2000;11:413–6. 153. Sommers DK, Loots JM, Simpson SF, et al. Circulating met-enkephalin in trained athletes during rest,exhaustive treadmill exercise and marathon running. Eur J Clin Pharmacol. 1990;38(4):391–2. 154. Sommers DK, Simpson SF, Loots JM, et al. Effect of exercise on met-enkephalin in unfit and superfit individuals. Eur J Clin Pharmacol. 1989;37(4):399–400. 155. Stagg NJ, Mata HP, Ibrahim MM, et al. Regular exercise reverses sensory hypersensitivity in a rat neuropathic pain model: role of endogenous opioids. Anesthesiology. 2011;114(4):940–8. 156. Sylvén C, Eriksson B, Sheps DS, et al. Beta-­ endorphin but not metenkephalin counteracts adenosine-provoked angina pectoris-like pain. Neuroreport. 1996;7(12):1982–4. 157. Tashiro M, Itoh M, Fujimoto T, et al. Application of positron emission tomography to neuroimaging in sports sciences. Methods. 2008;45:300–6. 158. Thorén P, Floras JS, Hoffmann P, Seals DR. Endorphins and exercise: physiological mechanisms and clinical implications. Med Sci Sports Exerc. 1990;22(4):417–28. 159. Tour J, Lofgren M, Mannerkorpi K, et al. Gene to gen interactions regulate endogenous pain modulation in fibromyalgia patients and healthy controls-­ atagonistic effects between opioid and serotonin-­ related genes. Pain. 2017;158(7):1194–203. 160. Triplett-McBride NT, Mastro AM, McBride JM, et al. Plasma proenkephalin peptide F and human B cell responses to exercise stress in fit and unfit women. Peptides. 1998;19(4):731–8. 161. Tsou K, Brown S, Sanudo-Pena MC, et al. Immunohistochemical distribution ocannabinoid CB1 receptors in the rat central nervous system. Neuroscience. 1998;83:393–411. 162. Tsuchimochi H, McCord JL, Kaufman MP. Peripheral mu-opioid receptors attenuate the augmented exercise pressor reflex in rats with chronic femoral artery occlusion. Am J Physiol Heart Circ Physiol. 2010;299(2):H557–65. 163. Umeda M, Lee W, Marino CW, Hilliard SC. Influence of moderate intensity physical activity levels and gender on conditioned pain modulation. J Sports Sci. 2016;34:467–76. 164. Vadivelu N, Mitra S, Hines RL. Peripheral opioid receptor agonists for analgesia: a comprehensive review. J Opioid Manag. 2011;7:55–68. 165. Van den Bergh P, Rozing J, Nagelkerken L. Two opposing modes of action of beta-endorphin on lymphocyte function. Immunology. 1991;72(4):537–43. 166. Van Epps DE, Saland L. Beta-endorphin and met-enkephalin stimulate human peripheral blood mononuclear cell chemotaxis. J Immunol. 1984;132(6):3046–53. 2 Endogenous Opiates and Exercise-Related Hypoalgesia 167. Van Middelkoop M, Rubinstein SM, Verhagen AP. Exercise therapy for chronic nonspecific low-back pain. Best Pract Res Clin Rheumatol. 2010;24:193–204. 168. Van Oosterwijck J, Nijs J, Meeus M, et al. Pain inhibition and post-exertional malaise in myalgic encephalomyelitis chronic fatigue syndrome: an experimental study. J Intern Med. 2010;268(3):265–78. 169. Van Oosterwijck J, Nijs J, Meeus M, et al. Lack of endogenous pain inhibition during exercise in people with chronic whiplash associated disorders: an experimental study. J Pain. 2012;13(3):242–54. 170. Viru A, Tendzegolskis Z, Smirnova T. Changes of beta-endorphin level in blood during prolonged exercise. Endocrinol Exp. 1990;1–2:63–8. 171. Vissing J, Iwamoto GA, Fuchs IE, et al. Reflex control of glucoregulatory exercise responses 172. 173. 174. 175. 39 by group III and IV muscle afferents. Am J Phys. 1994;266(3):R824–30. Walberg-Rankin J, Franke WD, Gwazdauskas FC. Response of beta-endorphin and estradiol to resistance exercise in females during energy balance and energy restriction. Int J Sports Med. 1992;13(7):542–7. Wen T, Peng B, Pintar JE. The MOR-1 opioid receptor regulates glucose homeostasis by modulating insulin secretion. Mol Endocrinol. 2009;23(5):671–8. Zitnik RJ, Whiting NL, Elias JA. Glucocorticoid inhibition of interleukin-1-induced interleukin-6 production by human lung fibroblasts: evidence for transcriptional and post-transcriptional regulatory mechanisms. Am J Respir Cell Mol Biol. 1994;10(6):643–50. Zollner C, Stein C. Opioids. Handb Exp Pharmacol. 2007;177:31–63. 3 The Effect of Exercise on the Hypothalamic-PituitaryAdrenal Axis David H. St-Pierre and Denis Richard The HPA Axis Introduction Over the last decades, important discoveries have allowed exercise science to bloom as a research field. Practical applications in kinesiology influence a wide range of populations including individuals with diverse degrees of disabilities to high-performance athletes. Important advances include the optimization of training techniques, biomechanics, motor skills, periodization, and injury prevention. Sports psychology is another emerging discipline recognized to have a profound impact on active individuals in terms of adherence and compliance to a training program as well as physical improvements and raw performance. As for injury prevention, it is now generally accepted that physical activity must be performed in an equilibrated way in order to maximize the desire to pursue while reducing the risks of nonadherence, of non-compliance, and of developing psychological disorders. Since its discovery, the hypothalamic-pituitary-adrenal D. H. St-Pierre (*) Department of Exercise Science, Département des Sciences de l’activité Physique, Université du Québec à Montréal (UQAM), Montréal, QC, Canada e-mail: st-pierre.david_h@uqam.ca D. Richard Quebec Heart and Lung Institute Research Center, Laval University Obesity Research Chair, Québec, QC, Canada (HPA) axis was shown to play a major role in the control of anxiogenic and depressive behaviors. A growing evidence indicate that exercise exerts acute and chronic effects on the HPA axis. However, the mechanisms through which it influences the HPA axis, and vice versa, remain to be clarified. To add to the complexity, a wide range of HPA axis responses are reported in different populations. These are generally proposed to depend on the type of physical activity, the intensity, and the volume at which it is achieved. Hence, overtraining and the dynamic progression of performance could also influence the relationship between exercise and the HPA axis. The present chapter will review the current state of knowledge to clarify how exercise influences the HPA axis. Defining the HPA Axis The HPA axis consists of three structurally independent components including the hypothalamus, the anterior pituitary, and the adrenal cortex (see Fig. 3.1). These structures are intimately interacting through the release of neuroendocrine messengers. In the medial parvocellular and the magnocellular parts of the paraventricular nucleus of the hypothalamus (PVH), corticotropin-­releasing factor [CRF, a 41-amino acid (aa) peptide] and arginine vasopressin (AVP, expressed in approximately half of the CRF neurons) are synthesized [1]. CRF neurons project © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_3 41 D. H. St-Pierre and D. Richard 42 Exercise (+) Ghrelin PACAP (-) Hypothalamus Stress Pituitary (+) CRF (+) BDNF (+) AVP IL-1b, IL-1, IL-6, and TNF-a Hypothalamus (+) (-) AVP CRF (+) Portal system Anterior pituitary Immune cells (+) Hypothalamic neurons Metabolic effects Corticotrophic cells Blood circulation (-) ACTH Systemic circulation Endotoxins (+) NOS GABA serotonin (+) Cortex Medulla Glucocorticoids (+) ACTH Receptors (MC2R) Adrenal gland Fig. 3.1 The three structurally independent components of the hypothalamic-pituitary-adrenal (HPA) which include the hypothalamus, the anterior pituitary, and the adrenal cortex to the exterior layer of the median eminence and release CRF into the portal circulation until they subsequently reach corticotroph cells from the anterior pituitary to stimulate the secretion of adrenocorticotropic hormone (ACTH). In turn, ACTH is released and transported via the general circulation to activate the adrenal secretion of glucocorticoids. Importantly, it is known that glucocorticoids negatively control pituitary corticotrophs and PVH CRF neurons through direct or hippocampus-mediated feedback inhibition mechanisms [2, 3]. In mammals, the CRF system is not limited to PVH CRF neurons. The system also comprises two CRF receptor types (CRF-R1 and CRF-R2) [4], a CRF-binding protein [5] and endogenous CRF receptor ligands, that include mammalian peptides CRF [6], urocortin (UCN) [7], UCN II [8, 9], and UCN III [9, 10]. In the brain, the broad distribution of CRFergic cells, UCNergic neurons, and CRF receptors is compatible with the main functions attributed to the CRF system [11]. Central administration of CRF evokes autonomic responses [2, 3], general arousal [12], as well as anxiety-like behaviors [3, 13]. Furthermore, central CRF injections also activate the sympathetic while inhibiting the parasympathetic branches of the autonomic nervous system by stimulating cardiorespiratory functions [14] and reducing the activity of the digestive system [15]. Because of their selectivity for CRF-R2, UCN II and UCN III [10] (also referred to stresscopin in humans) have been described as “stress-coping” peptides capable of exerting anxiolytic effects [9]. AVP is a 9-amino acid (aa) peptide with a disulfide bridge that is mainly secreted from the magnocellular cells of the supraoptic nucleus and the PVH and transported to the circulation to 3 The Effect of Exercise on the Hypothalamic-Pituitary-Adrenal Axis exert its effects on kidneys and blood vessels [16, 17]. In addition, AVP’s expression is also reported in the parvocellular neurons of the bed nucleus of the stria terminalis, the medial amygdala, the suprachiasmatic nucleus, and the PVH [18–20]. Three major types of AVP receptors are known: AVPR1a, AVPR1b, and AVPR2 [21, 22]. The activation of AVPR1b in the anterior pituitary stimulates the release of ACTH [23], while AVPR1a and AVPR2 are mainly expressed in the kidneys and blood vessels [24]. ACTH is a 39 aa peptide derived from the proteolytic cleavage of the proopiomelanocortin (POMC) gene [25–27]. The expression of ACTH is modulated positively by CRF and AVP, naloxone, interleukins (IL) IL-1 and IL-6, as well as leukemia inhibitory factor (LIF), but negatively by glucocorticoids [28–32]. However, other factors such as pituitary adenylate cyclase-activating peptide (PACAP), catecholamines, ghrelin, nitric oxide synthase (NOS), dihydroxyphenylalanine (DOPA), serotonin, and γ-aminobutyric acid (GABA) are also suspected to influence ACTH secretion through still ill-defined mechanisms [33–35]. ACTH is released in a pulsatile manner and has been shown to be regulated through a calcium-dependent mechanism [36]. It is subsequently transported in the circulation to activate the melanocortin type 2 receptor (MC2R) from the adrenal glands [37, 38] and, ultimately, stimulate species-specific glucocorticoid (either cortisol in human, nonhuman primates, pigs, and dogs or corticosterone in laboratory rodents such as rats and mice) synthesis and secretion [39]. In a matter of seconds to minutes, the release of glucocorticoids from adrenal glands will activate glucocorticoid receptors (GR), stimulate annexin 1 (ANXA1) production, and, consequently, block CRF-induced ACTH secretion [40, 41]. It is however suggested that the level of complexity of the direct and indirect mechanisms through which glucocorticoids exert their repressive effects on the HPA is much higher than what was anticipated during the 1980s [1]. Other mediators of the HPA axis were identified over the last decades. For instance, the gut microbiota is now proposed to influence anxiogenic and depressive behaviors via its effects on 43 the HPA axis. Germ-free (absence of gut microbiota) chronically restrained mice display antianxiety behaviors but increased CRF, ACTH, cortisol, and aldosterone levels in hypothalamic tissues compared to specific pathogen-free microbiota mice [42–45]. Although the microbial mechanisms influencing these effects remain ill-­ defined, it is proposed to regulate glucocorticoid receptor sensitivity (Fkbp5), steroidogenesis (MC2R, StaR, Cyp11a1), and catecholamine synthesis (TH, PNMT) [46]. Hence, colon expression of 11-β hydroxysteroid dehydrogenase 1 (11HSD-1), CRF, urocortin II and its receptor, and CRFR2 as well as cytokines TNFα, INFγ, IL-4, IL-5, IL-6, IL-10, IL-13, and IL-17 is also reported to be modulated by the microbiota. As recently evidenced, there is an intimate link between the regulation of the HPA axis and inflammatory cytokines [47]. For instance, interleukin 1β (IL-1β) is reported to influence the release of CRF in the hypothalamus, ACTH in the pituitary, and glucocorticoids in the adrenal cortex [48–53]. It was also reviewed that IL-6 and TNF-α promote the activation of the HPA axis [54]. Some of these effects are mediated through the activation of cyclooxygenase enzymes (prostaglandins) as well as by brain nitric oxide, noradrenaline, and serotonin production [55]. Interestingly, the translocation of endotoxins (derived from Gram-negative microbial components such as lipopolysaccharides/ LPS and others) was previously shown to activate the HPA axis through the release of IL-1, IL-6, and TNF-α [56]. This reinforces the existence of an intimate relation between the gut (and the microbiota) and the brain for the regulation of the HPA axis. Brain-derived neurotrophic factor (BDNF) is another factor with an influence on the HPA axis. For example, a single bout of exercise was shown to stimulate hippocampal BDNF expression in mice [57]. In humans, carriers of the Val66Met BDNF allele (prevalence of up to 50% and 32% in Asians and Caucasians, respectively [58]) were shown to display increased HPA axis activity through a higher cortisol response to stress [59, 60]. Expression of BDNF is co-localized with CRF and AVP in the PVH and the lateral 44 ventricle [61]. Hence, BDNF administrations increased the expression of CRF while exerting the opposite effect on AVP in the parvocellular and magnocellular PVH portions. Hence this treatment was likely to promote CRF secretion since its levels were decreased, while those of AVP were higher in the hypothalamus. This hypothesis is supported by the fact that the administration of BDNF also upregulated ACTH and corticosterone plasma concentrations. The HPA Axis and Exercise Endurance Training The effect of endurance training on the activation of the HPA axis has been investigated extensively in animal and human models. In pigs submitted to a high-fat diet, a 200% increase in free fatty acid (FFA) levels is related to a 40% decrease in ACTH concentrations in response to stress [62]. In the same study, pigs submitted to an endurance training program displayed a 60% increase in ACTH following a stress challenge; this effect was associated with a 56% decrease in FFA without other changes in body composition and insulin sensitivity. In another study, rats confined to a cage that allowed voluntary wheel running, corticosterone responses to various stimulatory challenges of the HPA axis were shown to be significantly higher than in untrained animals [63, 64]. Interestingly, this enhanced adrenal sensitivity to ACTH was completely restored to normal following 5–8 weeks of exercise training. In an ovine model, ACTH levels were found to rise in response to exercise, even though the animals had been previously submitted to a CRF infusion [65]. The latter suggests that ACTH release could be stimulated by other factor than CRF, and the authors suggested AVP as a plausible candidate. Endurance training upregulated mRNA expression of BDNF and its receptor TrkB in the hippocampus, midbrain, and striatum while increasing BDNF levels in the hippocampus and striatum in rats [66]. On the other hand, sprint interval training was more effective to enhance BDNF brain content than intensive endurance D. H. St-Pierre and D. Richard training in rats [67]. These increased BDNF levels in the brain were also shown to be associated with reduced anxiety- and depression-like behaviors in tested animals. In human studies, the activation of the HPA axis in response to physical activity has been abundantly reported. For instance, individuals submitted to chronic endurance training displayed higher hair cortisol [68]. In endurance-­ trained men, after a day without physical exercise, ACTH and cortisol concentrations were similar to those of untrained controls [69]. For most of these athletes, dexamethasone (a synthetic agonist of the glucocorticoid receptor) was not found to influence the activity of the HPA axis; however, in contrast to untrained subjects, a subsequent administration of CRF was shown to increase cortisol levels. On the other hand, obese adolescents submitted to a chronic physical activity program displayed a marginal decrease in glucocorticoid sensitivity and increased levels of glucocorticoid receptor-α (GR-α) expression in blood mononuclear cells [70]. In young men who were previously undergoing a strength training program, cortisol responses were significantly increased when submitted to higher frequencies of endurance training [71]. Twenty weeks of endurance training were also shown to decrease basal cortisol levels [72]. Hence, the magnitude of the reduction in cortisol levels was significantly associated with increases in local skeletal muscle endurance. As observed in animals, endurance training also significantly upregulated basal BDNF circulating levels in healthy sedentary or physically active males, and the authors suggest that this effect could promote brain health in these populations [73, 74]. The influence of an acute bout of endurance exercise on HPA axis activity has also been investigated in a multitude of studies. In response to a walk on a treadmill until exhaustion at 40 °C, circulating levels of cortisol were higher in trained than in untrained individuals, while those of ACTH were not different [75]. Interestingly, in response to the same challenge in trained and untrained individuals, ACTH, norepinephrine, and dehydroepiandrosterone-sulfate (DHEA-S) levels were significantly increased, while those 3 The Effect of Exercise on the Hypothalamic-Pituitary-Adrenal Axis of growth hormones (GHs), aldosterone and epinephrine, were initially elevated but reached a maximal value (plateau) at 38.5 °C. In athletes submitted to a strenuous exercise, CRF and cortisol responses to HPA activation were not blunted by physiological endogenous hypercortisolism, and this suggests that pituitary sensitivity is decreased in response to the feedback inhibition induced by cortisol [76]. As noted, acute physical activity has been reported to influence HPA axis activity; however, the relevance of considering other physiological conditions should not be neglected. In fasting subjects submitted to physical exhaustion, ACTH and cortisol levels significantly increased in hypoglycemic conditions, but this effect was abolished when pretest glycemic levels were maintained [77]. This also suggests the relevance of further examining the HPA axis activation under hypoglycemia. While the abovementioned information indicates that HPA axis activity is modulated by chronic and acute training, it is also important to evaluate the effect of a recuperation phase. In runners, it has been observed that cortisol and ACTH levels are significantly lower 2 days following a marathon, while whole body 11β-­HSD-­1 and ghrelin levels are upregulated [78]. Also, the suppression of cortisol in response to a dexamethasone challenge is strongly increased after 6 weeks of reduced training. Resistance Training Although the effect of endurance training on the activation of the HPA axis is abundantly described, fewer studies have evaluated the effect of resistance training. Resistance training can be defined as any exercise program using one or multiple training strategies (own body mass, free weights, or diverse exercising machines), to enhance health, fitness, and performance [79]. In healthy untrained men submitted to acute resistance training, cortisol concentrations were not modulated [80]. However, in the same subjects, catecholamines, lactate, TNF-α, IL-2, and epidermal growth factor (EGF) levels increased, while monocyte che- 45 motactic protein-1 (MCP-1) concentrations decreased. Furthermore, a positive correlation was observed between the concentrations of cortisol and TNF-α. Interestingly, the type and the intensity at which resistance training is performed are suggested to influence the HPA axis. In competitive athletes performing in muscular power disciplines (alpine ski, bodybuilding, and volleyball), an isokinetic exercise induced higher acute increases in ACTH, cortisol, and lactate than in endurance athletes (marathon, triathlon, cross-country skiing, and rowing) [81]. However, this effect was not observed during the recovery period. The type of training is reported to influence the activation of the HPA axis; however the effects of the intensity and volume of resistance training needed to be clarified. Interestingly, significantly lower cortisol levels were measured after a single bout of high-­intensity resistance training (HIT) then after performing a traditional 3-set protocol in male college students [82]. Age, gender, circadian rhythm, and body composition are other factors that are often reported to influence hormonal secretions (see Copeland, Chap. 23 in this book). Studies were conducted to clarify the effects of age, gender, circadian rhythm, and body composition on the activation of the HPA axis. Young and middle-aged men were submitted to an 8-week resistance training program which was shown to decrease both basal cortisol and ACTH levels [83]. However, age did not have a significant influence on the results. In contrast, 9 weeks of combined endurance and resistance training was shown to increase cortisol levels by 23% in young sedentary women, but this effect was not observed in their male counterparts [84]. This suggested that women undergoing physical training are more sensitive to the activation of the HPA axis than males. To determine the role of the circadian rhythm on the activation of the HPA axis, trained subjects were instructed to perform the same resistance training session at three time periods over different days. Cortisol levels were higher in the morning but decreased 3 min and up to 48 h after performing their bout of exercise [85]. This indicated the importance of considering the time at which blood samples are collected before, during, and 46 after undergoing a session of resistance training. Contrastingly, after submitting untrained young males to 11 weeks of resistance training, the time of the day at which exercises were performed did not influence the levels of hormones of the HPA axis [86]. However, the same authors reported that postexercise cortisol levels were lower than basal concentrations. To determine the effects of body composition on the activation of the HPA axis, normal weight and obese individuals were submitted to resistance training. Cortisol levels were significantly different between normal weight and obese individuals [87]. This suggested that body composition may also modulate the HPA axis. Different types of resistance training promote skeletal muscle hypertrophy or strength. Untrained young male and female adults were recruited to clarify the different effects of the two types of resistance training on the HPA axis. While performing the experimental protocol, significantly higher BDNF levels were measured during the exercise designed to promote hypertrophy than the one intended to increase strength [88]. In trained men, BDNF levels increased similarly in response to the different intensity and volume levels of resistance training [89]. In older adults submitted to various loads of resistance training, BDNF levels increased in male participants, while no effects could be detected in female individuals [90]. These data support the hypothesis that the HPA axis activation might be influenced by the type of training, the intensity, the post-training period, and body mass but not by age or the time of the day at which it is performed. These latter issues are in need of further investigations to clarify aspects of the contradictory results. I ntensity of Physical Activity and HPA Axis Activation It is profusely reported that the HPA axis is activated in response to physical activity, and different levels of exercise intensity were also shown to have an important impact. In mice submitted to acute psychological stress, high-intensity physi- D. H. St-Pierre and D. Richard cal activity increased cortisol, IL-1β, IL-2, and IL-6 while decreasing ACTH-positive cells in the pituitary [91]. Although collected in animals, these results indicated the relevance of considering the intensity of physical activity, and this was also investigated in human models. For instance, it was initially proposed that cortisol levels are increased by 60 min of running on a treadmill at a threshold intensity of 60% of the VO2max [92]. Moderately trained men also displayed a significant increase in cortisol after performing 30 min of exercise at 60% and 80% of their VO2max, while ACTH levels were only elevated at the highest intensity [93]. In endurance-trained males, 30 min of exercise on a cycle ergometer, significant increases in cortisol were only observed at 80% of the VO2max both in saliva serum [94]. Interestingly, the same authors observed that peak cortisol levels were only monitored 30 min after the cessation of the physical activity. When compared to low-intensity, high-intensity cycling caused similar increases in BDNF and cortisol levels in both participants with or without depression [95]. In trained athletes submitted to a prolonged high-intensity exercise, increased plasma concentrations of cortisol, ACTH, CRF, and AVP were observed [96]. It was also reported that the rise in osmolality observed during exercise correlates with increases in plasma AVP. Furthermore, for a given type of physical activity, high-­intensity and prolonged duration respectively increased AVP and CRF levels. In healthy participants administered with dexamethasone (4 mg), performing physical activity at the highest intensity (90% vs. 100% maximal aerobic capacity) caused a significant raise in ACTH, cortisol, and AVP circulating levels [97]. Interestingly, this response was shown to be amplified in women with regard to the one observed in men. Interestingly, high-­ intensity interval training was shown to increase BDNF levels to a higher magnitude than continuous moderate-intensity exercises in obese individuals [98]. This suggests that short and intense bouts of exercise could exert beneficial effects to individuals intending to design and/or perform physical activity programs. While the effect of exercise intensity was evaluated in response to distinct physical activities, 3 The Effect of Exercise on the Hypothalamic-Pituitary-Adrenal Axis another group compared occupational differences between workers performing high-­intensity duties (slaughterhouse workers) and others achieving low-intensity tasks (office workers) [99]. Slaughterhouse workers displayed higher levels of ACTH, total peroxides, antioxidant capacity, oxidative stress index, and c-reactive protein (CRP), while their levels of endogenous peroxidase activity, polyphenols, and BDNF were reduced. These results were even affected by the duration of the work shifts in slaughterhouse workers since higher CRP and lower BDNF levels were measured after completing 12 h vs. 8 h shifts. Results presented in this section clearly indicate that the intensity and the volume of a physical activity, the fitness level, and the type of exercise performed by an individual have a direct impact on the activation of the HPA axis. In turn, this should be taken into consideration when elaborating training programs. Highly Trained and Elite Athletes Overall, the increased activity of the HPA axis in highly trained athletes could have important implications on their somatic and mental health. During a progressive stress test until exhaustion on a treadmill, cortisol levels were higher from baseline to the initiation of recuperation in professional athletes than in controls [100]. Interestingly, hormonal levels were regularized over the recuperation period. In ultramarathon runners, cortisol levels were at their highest at the completion of a 622 km race, and levels were only normalized after 6 days of recovery [101]. In highly trained athletes, the morning surge in ACTH and cortisol was observed earlier, and ACTH levels were significantly higher than in normal individuals [102]. In addition, the stimulation of CRF and ACTH release was more pronounced in highly trained athletes than in untrained individuals following the administration of the nonselective opioid receptor antagonist naloxone [32]. Altered HPA axis functions were also observed in elite athletes. For instance, artistic gymnasts competing at the European 47 Championships displayed higher salivary cortisol concentrations and more important levels of psychological stress than controls [103]. In addition, higher psychological stress and saliva cortisol levels were also observed in female vs. male athletes. In elite junior soccer players, nonfunctional overreaching performances were associated with higher scores of depression and angriness, whereas resting GH and ACTH concentrations after maximal effort were diminished [104]. These observations could be associated with the decreased expression of GR-α mRNA in highly trained individuals and with lower increases in atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) levels in response to exercise [105–110]. These elements suggest the influence of the HPA axis on stress and emotional status. Ultimately these factors could also have a major incidence on sportive performances in elite athletes. Overtraining The available information regarding altered HPA axis functions in athletes suggests the relevance of considering potentially for pathological conditions such as overtraining. In rats submitted to daily swimming bouts of 45 min 5 days per week for 2, 4, or 6 weeks, corticosterone gradually increased. In parallel both basal ACTH and corticosterone plasma levels increased until they reached a plateau after 6 weeks of swimming [111]. Hence, in the PVN and the pituitary of the same animals, mRNA expression of the glucocorticoid receptor decreased, while the one of CRF transiently increased. While these results are interesting, it is difficult to determine whether the important volume of exercise to which rats were submitted can be considered as overtraining. These results raise important questions since cortisol levels were significantly below normal in overtrained Standardbred racehorses [112, 113]. Interestingly, these discrepancies may be species-­ specific or be related to the duration overtraining in the animals. In other words, rats submitted to swimming may still have the capacity to produce corticosterone, while Standardbred D. H. St-Pierre and D. Richard 48 racehorses may have been submitted to a chronic overactivation of the HPA axis which led to impairments in their capacities to secrete cortisol before being diagnosed. In different populations of human athletes, several alternative methods such as a CRF stimulation test (evaluation of basal ACTH concentrations and GH pulsatility), free testosterone over cortisol concentrations, low basal cortisol levels, as well as HPA responses to two standardized exercise tests were proposed for the diagnostic of overtraining [114–116]. For instance, in response to two acute bouts of exercise, increased prolactin (PRL) levels and decreased ACTH concentrations are reported in overtrained athletes [117– 119]. These effects could be mediated by the repetitive occurrence of muscle and skeletal trauma resulting in local inflammation and, consequently, in a systemic inflammatory responses which, in turn, could yield to impairments of athletic performances [115]. Postexercise Recuperation Depending on the type of physical activity and its intensity and volume, it is critical to allow the body to recuperate, replenish its energy reserves, and resynthesize injured tissues in order to improve athletic performance. Recuperation is well-characterized in nutrition and physiology; however, it is another factor to take into consideration when considering the effects of exercise on the HPA axis. For instance, it was shown that the carbohydrate/electrolyte consumption right after performing a bout of high-intensity physical activity significantly reduced blood cortisol levels in male athletes [120]. However, the hydration status, per se, was not associated with an alteration of circulating cortisol concentrations [121]. In rugby players submitted to a magnesium supplementation, significantly higher ACTH but decreased cortisol levels were observed compared to the same type of participants given a placebo [122]. In addition, magnesium supplementation abolished the post-game increase in IL-6 while reducing the increase in neutrophil/lymphocyte ratio. emory, Defeat, Fear, and Cognitive M Functions During a physical activity, the capacity to remember how to optimally perform an exercise as well as the bad feelings and the fear of defeat or mishaps occurring during the event may have profound effects on an individual’s performance. Because of obvious ethical reasons, this is difficult to investigate in humans. However, rodent models were used to investigate the effect of the HPA axis on memory, defeat, and fear. In rats administered with metyrapone (a corticosterone synthesis disruptor), impaired traces of fear conditioning have been observed [123]. A number of studies also evaluated the effects of CRF on defeat conditioning as well as on memory. The central administration of anti-sauvagine-30 (a CRF-R2 receptor antagonist) reduced submissive and defensive behaviors induced by territorial aggression conditioning in Syrian hamsters [124]. However this effect was not observed in response to neither metyrapone nor CP-154,526 (a CRF-R1 antagonist) administrations in the lateral ventricle of rats increased spatial memory through a β-adrenergic-dependent mechanism [125]. Also, central administrations of NBI30775 (CRF-R1 antagonist) prevented stress-induced hippocampal dendritic spine loss while restoring stress-impaired cognitive functions [126]. This suggests that stress-induced central effects are mediated through the activation of CRF-R1. These discoveries are important since they could allow the implementation of targeted interventions or pharmacological treatments to reduce the fear of defeat or the occurring of an injury in individuals or athletes who previously encountered such negative experiences. In turn, this would allow preventing the adverse outcomes on their athletic performances. Conclusion The last paragraphs have underlined the importance of the HPA axis on the regulation of moods and behaviors in animals and humans undergoing physical activity. Depending on the population of 3 The Effect of Exercise on the Hypothalamic-Pituitary-Adrenal Axis interest and the objectives to be reached, it is critical to adapt training programs to maximize their benefits while minimizing the risk of developing anxiety and depression. This has to be applied to athletes as well as other populations with different levels of fitness and/or degrees of disabilities. For instance, professional or Olympic athletes are particularly at risk of overtraining, and their moods, cognitive functions, and confidence levels have an important impact on their performances. Physical activity is also an important element of a healthy lifestyle to prevent and/ or counteract the dreadful effects of obesity and ensuing metabolic dysfunctions that have reached epidemic levels in North American populations. In obese individuals, adherence and compliance to training programs remain major obstacles. For athletes or obese individuals, a better understanding on how exercise modulates the HPA axis will provide essential tools to develop novel training approaches. As one consequence of this, it will be essential to exhaustively characterize individuals undergoing an exercise program in order to determine their levels of fitness; the type, intensity, and volume of physical activity required; as well as the window of time over which objectives need to be achieved. It is also important to constantly monitor exercising individuals since adaptation to the training planification will be required as soon as anxiogenic or depressive behaviors will be present. Hence, distinct factors of the HPA axis may be used as sensitive biomarkers to detect disorders before clinical symptoms can be detected. Globally, this indicates the relevance of including parameters of the HPA axis as modulators of anxiety and depressive behaviors in exercising individuals. In sum, while it remains a precarious equilibrium, it suggests that elements of the HPA axis must be taken into consideration along with the assessment of an individual’s physical capacities when designing a training program. Consequently, while the planification and periodization must be optimized, it is important to adapt the program accordingly in function of early signs of anxiogenic or depressive behaviors. Ultimately, this will be more effective at yielding improvements in athletic performance and health benefits than the simple addition of 49 strenuous exercises that could provoke the premature interruption or to slow down physical training program for various clinical reasons. Our current knowledge of the relationship between the HPA axis and physical exercise reviewed in the above paragraphs clearly highlights the importance of adequate preparation for exercise. Also, a number of data indicate that complex molecular and cellular mechanisms intervene in highly trained athletes that do not occur in normal individuals. As a whole, the above information suggests that the HPA axis importantly influences stress-induced functions and that the intensity of HPA axis activation is intimately related to the type of training, the intensity, and the volume at which it is performed. This strongly suggests the relevance of considering the impact of the HPA axis when elaborating training programs in different types of individuals. References 1. Watts AG. Glucocorticoid regulation of peptide genes in neuroendocrine CRH neurons: a complexity beyond negative feedback. Front Neuroendocrinol. 2005;26:109–30. https://doi. org/10.1016/j.yfrne.2005.09.001. 2. Brown MR, Fisher LA. In: Nemeroff CB, De Souza EB, editors. Corticotropin-releasing factor: basic and clinical studies of a neuropeptide. CRC. Taylor & Francis group 1990. p. 291–8. 3. Heinrichs SC, Tache Y. Therapeutic potential of CRF receptor antagonists: a gut-brain perspective. Expert Opin Investig Drugs. 2001;10:647–59. https://doi. org/10.1517/13543784.10.4.647. 4. Muller MB, Wurst W. Getting closer to affective disorders: the role of CRH receptor systems. Trends Mol Med. 2004;10:409–15. https://doi. org/10.1016/j.molmed.2004.06.007. 5. Linton EA, Behan DP, Saphier PW, Lowry PJ. Corticotropin-releasing hormone (CRH)-binding protein: reduction in the adrenocorticotropin-­ releasing activity of placental but not hypothalamic CRH. J Clin Endocrinol Metab. 1990;70:1574–80. 6. Vale W, Spiess J, Rivier C, Rivier J. Characterization of a 41-residue ovine hypothalamic peptide that stimulates secretion of corticotropin and beta-­ endorphin. Science. 1981;213:1394–7. 7. Vaughan J, et al. Urocortin, a mammalian neuropeptide related to fish urotensin I and to corticotropin-­ releasing factor. Nature. 1995;378:287–92. https:// doi.org/10.1038/378287a0. D. H. St-Pierre and D. Richard 50 8. Reyes TM, et al. Urocortin II: a member of the corticotropin-­ releasing factor (CRF) neuropeptide family that is selectively bound by type 2 CRF receptors. Proc Natl Acad Sci U S A. 2001;98:2843– 8. https://doi.org/10.1073/pnas.051626398. 9. Hsu SY, Hsueh AJ. Human stresscopin and stresscopin-­ related peptide are selective ligands for the type 2 corticotropin-releasing hormone receptor. Nat Med. 2001;7:605–11. https://doi. org/10.1038/87936. 10. Lewis K, et al. Identification of urocortin III, an additional member of the corticotropin-releasing factor (CRF) family with high affinity for the CRF2 receptor. Proc Natl Acad Sci U S A. 2001;98:7570– 5. https://doi.org/10.1073/pnas.121165198. 11. Turnbull AV, Rivier C. Corticotropin-releasing factor (CRF) and endocrine responses to stress: CRF receptors, binding protein, and related peptides. Proc Soc Exp Biol Med. 1997;215:1–10. 12. Koob GF, Cole BJ, Swerdlow NR, Le Moal M, Britton KTS. performance, and arousal: focus on CRF. NIDA Res Monogr. 1990;97:163–76. 13. Krysiak R, Obuchowicz E, Herman ZS. Role of corticotropin-releasing factor (CRF) in anxiety. Pol J Pharmacol. 2000;52:15–25. 14. Fisher LA, et al. Corticotropin-releasing factor (CRF): central effects on mean arterial pressure and heart rate in rats. Endocrinology. 1982;110:2222–4. 15. Taché Y, Gunion MM, Stephens R. In: Nemeroff CB, De Souza EB, editors. Corticotropin-releasing factor: basic and clinical studies of a neuropeptide. CRC. Taylor & Francis group 1990. p. 299–307. 16. Brownstein MJ, Russell JT, Gainer H. Synthesis, transport, and release of posterior pituitary hormones. Science. 1980;207:373–8. 17. Nishimura H, Fan Z. Regulation of water movement across vertebrate renal tubules. Comp Biochem Physiol A Mol Integr Physiol. 2003;136:479–98. 18. Dogterom J, Snijdewint FG, Buijs RM. The distribution of vasopressin and oxytocin in the rat brain. Neurosci Lett. 1978;9:341–6. 19. Buijs RM. Intra- and extrahypothalamic vasopressin and oxytocin pathways in the rat. Pathways to the limbic system, medulla oblongata and spinal cord. Cell Tissue Res. 1978;192:423–35. 20. DeVries GJ, Buijs RM, Van Leeuwen FW, Caffe AR, Swaab DF. The vasopressinergic innervation of the brain in normal and castrated rats. J Comp Neurol. 1985;233:236–54. https://doi.org/10.1002/ cne.902330206. 21. Michell RH, Kirk CJ, Billah MM. Hormonal stimulation of phosphatidylinositol breakdown with particular reference to the hepatic effects of vasopressin. Biochem Soc Trans. 1979;7:861–5. 22. Jard S, Lombard C, Marie J, Devilliers G. Vasopressin receptors from cultured mesangial cells resemble V1a type. Am J Phys. 1987;253:F41–9. 23. Antoni FA, Holmes MC, Makara GB, Karteszi M, Laszlo FA. Evidence that the effects of arginine-­ 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 8-­ vasopressin (AVP) on pituitary corticotropin (ACTH) release are mediated by a novel type of receptor. Peptides. 1984;5:519–22. Bankir L. Antidiuretic action of vasopressin: quantitative aspects and interaction between V1a and V2 receptor-mediated effects. Cardiovasc Res. 2001;51:372–90. Bicknell AB. The tissue-specific processing of pro-­ opiomelanocortin. J Neuroendocrinol. 2008;20:692–9. https://doi. org/10.1111/j.1365-2826.2008.01709.x. Oliver RL, Davis JR, White A. Characterisation of ACTH related peptides in ectopic Cushing’s syndrome. Pituitary. 2003;6:119–26. Raffin-Sanson ML, de Keyzer Y, Bertagna X. Proopiomelanocortin, a polypeptide precursor with multiple functions: from physiology to pathological conditions. Eur J Endocrinol. 2003;149:79–90. Papadimitriou A, Priftis KN. Regulation of the hypothalamic-pituitary-adrenal axis. Neuroimmunomodulation. 2009;16:265–71. https:// doi.org/10.1159/000216184. Itoi K, Jiang YQ, Iwasaki Y, Watson SJ. Regulatory mechanisms of corticotropin-releasing hormone and vasopressin gene expression in the hypothalamus. J Neuroendocrinol. 2004;16:348–55. https://doi. org/10.1111/j.0953-8194.2004.01172.x. Mastorakos G, Chrousos GP, Weber JS. Recombinant interleukin-6 activates the hypothalamic-pituitary-­ adrenal axis in humans. J Clin Endocrinol Metab. 1993;77:1690–4. Crofford LJ, et al. Circadian relationships between interleukin (IL)-6 and hypothalamic-pituitary-­ adrenal axis hormones: failure of IL-6 to cause sustained hypercortisolism in patients with early untreated rheumatoid arthritis. J Clin Endocrinol Metab. 1997;82:1279–83. Inder WJ, et al. Elevated basal adrenocorticotropin and evidence for increased central opioid tone in highly trained male athletes. J Clin Endocrinol Metab. 1995;80:244–8. White A. Adrenocorticotropic hormone. Endocrinology. Publisher Elsevier Saunders; 2005:323–39. Arvat E, et al. Endocrine activities of ghrelin, a natural growth hormone secretagogue (GHS), in humans: comparison and interactions with hexarelin, a nonnatural peptidyl GHS, and GH-releasing hormone. J Clin Endocrinol Metab. 2001;86:1169–74. Jankord R, McAllister RM, Ganjam VK, Laughlin MH. Chronic inhibition of nitric oxide synthase augments the ACTH response to exercise. Am J Physiol Regul Integr Comp Physiol. 2009;296:R728–34. https://doi.org/10.1152/ajpregu.90709.2008. Gambacciani M, et al. Intrinsic pulsatility of ACTH release from the human pituitary in vitro. Clin Endocrinol. 1987;26:557–63. Xia Y, Wikberg JE. Localization of ACTH receptor mRNA by in situ hybridization in mouse adrenal gland. Cell Tissue Res. 1996;286:63–8. 3 The Effect of Exercise on the Hypothalamic-Pituitary-Adrenal Axis 38. Gorrigan RJ, Guasti L, King P, Clark AJ, Chan LF. Localisation of the melanocortin-2-receptor and its accessory proteins in the developing and adult adrenal gland. J Mol Endocrinol. 2011;46:227–32. https://doi.org/10.1530/JME-11-0011. 39. Chan LF, Metherell LA, Clark AJ. Effects of melanocortins on adrenal gland physiology. Eur J Pharmacol. 2011;660:171–80. https://doi. org/10.1016/j.ejphar.2010.11.041. 40. John CD, Gavins FN, Buss NA, Cover PO, Buckingham JC. Annexin A1 and the formyl peptide receptor family: neuroendocrine and metabolic aspects. Curr Opin Pharmacol. 2008;8:765–76. https://doi.org/10.1016/j.coph.2008.09.005. 41. Buckingham JC. Glucocorticoids: exemplars of multi-tasking. Br J Pharmacol. 2006;147(Suppl 1):S258–68. https://doi.org/10.1038/sj.bjp.0706456. 42. Crumeyrolle-Arias M, et al. Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology. 2014;42:207–17. https://doi.org/10.1016/j.psyneuen.2014.01.014. 43. Desbonnet L, Clarke G, Shanahan F, Dinan TG, Cryan JF. Microbiota is essential for social development in the mouse. Mol Psychiatry. 2014;19:146–8. https://doi.org/10.1038/mp.2013.65. 44. Wong ML, et al. Inflammasome signaling affects anxiety- and depressive-like behavior and gut microbiome composition. Mol Psychiatry. 2016;21:797– 805. https://doi.org/10.1038/mp.2016.46. 45. Huo R, et al. Microbiota modulate anxiety-­ like behavior and endocrine abnormalities in Hypothalamic-Pituitary-Adrenal axis. Front Cell Infect Microbiol. 2017;7:489. https://doi. org/10.3389/fcimb.2017.00489. 46. Vodicka M, et al. Microbiota affects the expression of genes involved in HPA axis regulation and local metabolism of glucocorticoids in chronic psychosocial stress. Brain Behav Immun. 2018;73:615–24. https://doi.org/10.1016/j.bbi.2018.07.007. 47. Papargyri P, et al. Links between HPA axis and adipokines: clinical implications in paradigms of stress-­ related disorders. Expert Rev Endocrinol Metab. 2018;13:317–32. https://doi.org/10.1080/17446651. 2018.1543585. 48. Goshen I, Yirmiya R. Interleukin-1 (IL-1): a central regulator of stress responses. Front Neuroendocrinol. 2009;30:30–45. https://doi.org/10.1016/j. yfrne.2008.10.001. 49. Mohn CE, et al. Adrenal gland responses to lipopolysaccharide after stress and ethanol administration in male rats. Stress. 2011;14:216–26. https:// doi.org/10.3109/10253890.2010.532254. 50. Gadek-Michalska A, Bugajski J. Interleukin-1 (IL-1) in stress-induced activation of limbic-­ hypothalamic-­ pituitary adrenal axis. Pharmacol Rep. 2010;62:969–82. 51. Gadek-Michalska A, Tadeusz J, Rachwalska P, Spyrka J, Bugajski J. Effect of repeated restraint on homotypic stress-induced nitric oxide synthases 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 51 expression in brain structures regulating HPA axis. Pharmacol Rep. 2012;64:1381–90. Gibb J, Hayley S, Poulter MO, Anisman H. Effects of stressors and immune activating agents on peripheral and central cytokines in mouse strains that differ in stressor responsivity. Brain Behav Immun. 2011;25:468–82. https://doi.org/10.1016/j. bbi.2010.11.008. Marquez C, Nadal R, Armario A. The hypothalamic-­ pituitary-­ adrenal and glucose responses to daily repeated immobilisation stress in rats: individual differences. Neuroscience. 2004;123:601–12. Dunn AJ. Cytokine activation of the HPA axis. Ann N Y Acad Sci. 2000;917:608–17. Gadek-Michalska A, Tadeusz J, Rachwalska P, Bugajski J. Cytokines, prostaglandins and nitric oxide in the regulation of stress-response systems. Pharmacol Rep. 2013;65:1655–62. Beishuizen A, Thijs LG. Endotoxin and the hypothalamo-pituitary-adrenal (HPA) axis. J Endotoxin Res. 2003;9:3–24. https://doi. org/10.1179/096805103125001298. Venezia AC, Quinlan E, Roth SM. A single bout of exercise increases hippocampal Bdnf: influence of chronic exercise and noradrenaline. Genes Brain Behav. 2017;16:800–11. https://doi.org/10.1111/ gbb.12394. Balkaya M, Cho S. Genetics of stroke recovery: BDNF val66met polymorphism in stroke recovery and its interaction with aging. Neurobiol Dis. 2018;126:36. https://doi.org/10.1016/j. nbd.2018.08.009. Jiang R, et al. Brain-derived neurotrophic factor (BDNF) Val66Met polymorphism interacts with gender to influence cortisol responses to mental stress. Psychoneuroendocrinology. 2017;79:13–9. https://doi.org/10.1016/j.psyneuen.2017.02.005. Schule C, et al. Brain-derived neurotrophic factor Val66Met polymorphism and dexamethasone/CRH test results in depressed patients. Psychoneuroendocrinology. 2006;31:1019–25. https://doi.org/10.1016/j.psyneuen.2006.06.002. Givalois L, et al. A single brain-derived neurotrophic factor injection modifies hypothalamo-­ pituitary-­ adrenocortical axis activity in adult male rats. Mol Cell Neurosci. 2004;27:280–95. https://doi. org/10.1016/j.mcn.2004.07.002. Jankord R, Ganjam VK, Turk JR, Hamilton MT, Laughlin MH. Exercise training alters effect of high-­ fat feeding on the ACTH stress response in pigs. Appl Physiol Nutr Metab. 2008;33:461–9. https:// doi.org/10.1139/H08-022. Campbell JE, Rakhshani N, Fediuc S, Bruni S, Riddell MC. Voluntary wheel running initially increases adrenal sensitivity to adrenocorticotrophic hormone, which is attenuated with long-term training. J Appl Physiol. 2009;106:66–72. https://doi. org/10.1152/japplphysiol.91128.2008. Fediuc S, Campbell JE, Riddell MC. Effect of voluntary wheel running on circadian c­orticosterone D. H. St-Pierre and D. Richard 52 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. release and on HPA axis responsiveness to restraint stress in Sprague-Dawley rats. J Appl Physiol. 2006;100:1867–75. https://doi.org/10.1152/ japplphysiol.01416.2005. Smoak B, Deuster P, Rabin D, Chrousos G. Corticotropin-releasing hormone is not the sole factor mediating exercise-induced adrenocorticotropin release in humans. J Clin Endocrinol Metab. 1991;73:302–6. Chalimoniuk M, Chrapusta SJ, Lukacova N, Langfort J. Endurance training upregulates the nitric oxide/soluble guanylyl cyclase/cyclic guanosine 3′,5′-monophosphate pathway in the striatum, midbrain and cerebellum of male rats. Brain Res. 2015;1618:29–40. https://doi.org/10.1016/j. brainres.2015.05.020. TaheriChadorneshin H, Cheragh-Birjandi S, Ramezani S, Abtahi-Eivary SH. Comparing sprint and endurance training on anxiety, depression and its relation with brain-derived neurotrophic factor in rats. Behav Brain Res. 2017;329:1–5. https://doi. org/10.1016/j.bbr.2017.04.034. Skoluda N, Dettenborn L, Stalder T, Kirschbaum C. Elevated hair cortisol concentrations in endurance athletes. Psychoneuroendocrinology. 2012;37:611– 7. https://doi.org/10.1016/j.psyneuen.2011.09.001. Duclos M, Corcuff JB, Pehourcq F, Tabarin A. Decreased pituitary sensitivity to glucocorticoids in endurance-trained men. Eur J Endocrinol. 2001;144:363–8. Faria CD, et al. Impact of prolonged low-grade physical training on the in vivo glucocorticoid sensitivity and on glucocorticoid receptor-alpha mRNA levels of obese adolescents. Horm Res Paediatr. 2010;73:458– 64. https://doi.org/10.1159/000313591. Jones TW, Howatson G, Russell M, French DN. Performance and endocrine responses to differing ratios of concurrent strength and endurance training. J Strength Cond Res. 2016;30:693–702. https://doi.org/10.1519/JSC.0000000000001135. Grandys M, et al. The importance of the training-­ induced decrease in basal cortisol concentration in the improvement in muscular performance in humans. Physiol Res. 2016;65:109–20. Seifert T, et al. Endurance training enhances BDNF release from the human brain. Am J Physiol Regul Integr Comp Physiol. 2010;298:R372–7. https://doi. org/10.1152/ajpregu.00525.2009. Zoladz JA, et al. Endurance training increases plasma brain-derived neurotrophic factor concentration in young healthy men. J Physiol Pharmacol. 2008;59(Suppl 7):119–32. Wright HE, Selkirk GA, McLellan TM. HPA and SAS responses to increasing core temperature during uncompensable exertional heat stress in trained and untrained males. Eur J Appl Physiol. 2010;108:987– 97. https://doi.org/10.1007/s00421-009-1294-0. Duclos M, et al. Corticotroph axis sensitivity after exercise in endurance-trained athletes. Clin Endocrinol. 1998;48:493–501. 77. Tabata I, Ogita F, Miyachi M, Shibayama H. Effect of low blood glucose on plasma CRF, ACTH, and cortisol during prolonged physical exercise. J Appl Physiol. 1991;71:1807–12. 78. Bobbert T, et al. Adaptation of the hypothalamic-­ pituitary hormones during intensive endurance training. Clin Endocrinol. 2005;63:530–6. https://doi. org/10.1111/j.1365-2265.2005.02377.x. 79. Ratel S. High-intensity and resistance training and elite young athletes. Med Sport Sci. 2011;56:84–96. https://doi.org/10.1159/000320635. 80. Fatouros I, et al. Acute resistance exercise results in catecholaminergic rather than hypothalamic-­ pituitary-­ adrenal axis stimulation during exercise in young men. Stress. 2010;13:461–8. https://doi. org/10.3109/10253891003743432. 81. Minetto MA, et al. Corticotroph axis sensitivity after exercise: comparison between elite athletes and sedentary subjects. J Endocrinol Invest. 2007;30:215–23. 82. Cintineo HP, et al. Acute physiological responses to an intensity-and time-under-tension-equated singlevs. multiple-set resistance training bout in trained men. J Strength Cond Res. 2018;32:3310–8. https:// doi.org/10.1519/JSC.0000000000002872. 83. Arazi H, Damirchi A, Asadi A. Age-related hormonal adaptations, muscle circumference and strength development with 8 weeks moderate intensity resistance training. Ann Endocrinol (Paris). 2013;74:30– 5. https://doi.org/10.1016/j.ando.2012.11.004. 84. Kyrolainen H, et al. Effects of combined strength and endurance training on physical performance and biomarkers of healthy young women. J Strength Cond Res. 2018;32:1554–61. https://doi.org/10.1519/ JSC.0000000000002034. 85. Ammar A, et al. Acute and delayed responses of steroidal hormones, blood lactate and biomarkers of muscle damage after a resistance training session: time-of-day effects. J Sports Med Phys Fitness. 2018;58:980–9. https://doi.org/10.23736/ S0022-4707.17.07048-7. 86. Sedliak M, et al. Morphological, molecular and hormonal adaptations to early morning versus afternoon resistance training. Chronobiol Int. 2018;35:450–64. https://doi.org/10.1080/07420528.2017.1411360. 87. Sheikholeslami-Vatani D, Ahmadi S, Salavati R. Comparison of the effects of resistance exercise orders on number of repetitions, serum IGF-1, testosterone and cortisol levels in normal-weight and obese men. Asian J Sports Med. 2016;7:e30503. https://doi.org/10.5812/asjsm.30503. 88. Marston KJ, et al. Intense resistance exercise increases peripheral brain-derived neurotrophic factor. J Sci Med Sport. 2017;20:899–903. https://doi. org/10.1016/j.jsams.2017.03.015. 89. Church DD, et al. Comparison of high-­ intensity vs. high-volume resistance training on the BDNF response to exercise. J Appl Physiol (1985). 2016;121:123–8. https://doi.org/10.1152/ japplphysiol.00233.2016. 3 The Effect of Exercise on the Hypothalamic-Pituitary-Adrenal Axis 90. Nuvagah Forti L, et al. High versus low load resistance training: the effect of 24 weeks detraining on serum Brain Derived-Neurotrophic Factor (BDNF) in older adults. J Frailty Aging. 2017;6:53–8. https:// doi.org/10.14283/jfa.2017.2. 91. Wang J, et al. The impact of water-floating and high-­ intensity exercise on rat’s HPA axis and interleukins concentrations. Acta Physiol Hung. 2012;99:261– 70. https://doi.org/10.1556/APhysiol.99.2012.3.3. 92. Davies CT, Few JD. Effects of exercise on adrenocortical function. J Appl Physiol. 1973;35:887–91. https://doi.org/10.1152/jappl.1973.35.6.887. 93. Hill EE, et al. Exercise and circulating cortisol levels: the intensity threshold effect. J Endocrinol Invest. 2008;31:587–91. https://doi.org/10.1007/ BF03345606. 94. VanBruggen MD, Hackney AC, McMurray RG, Ondrak KS. The relationship between serum and salivary cortisol levels in response to different intensities of exercise. Int J Sports Physiol Perform. 2011;6:396–407. 95. Ross RE, Saladin ME, George MS, Gregory CM. High-intensity aerobic exercise acutely increases brain-derived neurotrophic factor. Med Sci Sports Exerc. 2019;51(8):1698–709. https://doi. org/10.1249/MSS.0000000000001969. 96. Inder WJ, Hellemans J, Swanney MP, Prickett TC, Donald RA. Prolonged exercise increases peripheral plasma ACTH, CRH, and AVP in male athletes. J Appl Physiol. 1998;85:835–41. 97. Deuster PA, et al. High intensity exercise promotes escape of adrenocorticotropin and cortisol from suppression by dexamethasone: sexually dimorphic responses. J Clin Endocrinol Metab. 1998;83:3332– 8. https://doi.org/10.1210/jcem.83.9.5110. 98. Rodriguez AL, et al. Acute high-intensity interval exercise induces greater levels of serum brain-­ derived neurotrophic factor in obese individuals. Exp Biol Med (Maywood). 2018;243(14):1153–60. https://doi.org/10.1177/1535370218812191. 99. Zelzer S, et al. Work intensity, low-grade inflammation, and oxidative status: a comparison between office and slaughterhouse workers. Oxidative Med Cell Longev. 2018;2018:2737563. https://doi. org/10.1155/2018/2737563. 100. Popovic B, et al. Acute response to endurance exercise stress: focus on catabolic/anabolic interplay between cortisol, testosterone, and sex hormone binding globulin in professional athletes. J Med Biochem. 2019;38:6–12. https://doi.org/10.2478/ jomb-2018-0016. 101. Choi ES, et al. Changes in hormone levels of participants in a 622-km ultramarathon race based on distance and recovery period. J Sports Med Phys Fitness. 2018;59(4):700–7. https://doi.org/10.23736/ S0022-4707.18.08533-X. 102. Wittert GA, Livesey JH, Espiner EA, Donald RA. Adaptation of the hypothalamopituitary adrenal axis to chronic exercise stress in humans. Med Sci Sports Exerc. 1996;28:1015–9. 53 103. Georgopoulos NA, et al. Abolished circadian rhythm of salivary cortisol in elite artistic gymnasts. Steroids. 2011;76:353–7. https://doi.org/10.1016/j. steroids.2010.10.013. 104. Schmikli SL, de Vries WR, Brink MS, Backx FJ. Monitoring performance, pituitary-adrenal hormones and mood profiles: how to diagnose non-­ functional over-reaching in male elite junior soccer players. Br J Sports Med. 2012;46:1019. https://doi. org/10.1136/bjsports-2011-090492. 105. Wisen AG, Ekberg K, Wohlfart B, Ekman R, Westrin A. Plasma ANP and BNP during exercise in patients with major depressive disorder and in healthy controls. J Affect Disord. 2011;129:371–5. https://doi. org/10.1016/j.jad.2010.09.002. 106. Carr BR, Mason JI. The effects of alpha-human atrial natriuretic polypeptide on steroidogenesis by fetal zone cells of the human fetal adrenal gland. Am J Obstet Gynecol. 1988;159:1361–5. 107. Crandall ME, Gregg CM. In vitro evidence for an inhibitory effect of atrial natriuretic peptide on vasopressin release. Neuroendocrinology. 1986;44:439–45. 108. Strohle A, Holsboer F. Stress responsive neurohormones in depression and anxiety. Pharmacopsychiatry. 2003;36(Suppl 3):S207–14. https://doi.org/10.1055/s-2003-45132. 109. Strohle A, Kellner M, Holsboer F, Wiedemann K. Atrial natriuretic hormone decreases endocrine response to a combined dexamethasone-­ corticotropin-­ releasing hormone test. Biol Psychiatry. 1998;43:371–5. 110. Bonifazi M, et al. Glucocorticoid receptor mRNA expression in peripheral blood mononuclear cells in high trained compared to low trained athletes and untrained subjects. J Endocrinol Invest. 2009;32:816–20. https://doi.org/10.3275/6428. 111. Park E, et al. Changes in basal hypothalamo-­ pituitary-­ adrenal activity during exercise training are centrally mediated. Am J Physiol Regul Integr Comp Physiol. 2005;289:R1360–71. https://doi. org/10.1152/ajpregu.00103.2005. 112. de Graaf-Roelfsema E, Keizer HA, van Breda E, Wijnberg ID, van der Kolk JH. Hormonal responses to acute exercise, training and overtraining. A review with emphasis on the horse. Vet Q. 2007;29:82–101. 113. Cayado P, et al. Hormone response to training and competition in athletic horses. Equine Vet J Suppl. 2006;38:274–8. 114. Banfi G, Dolci A. Free testosterone/cortisol ratio in soccer: usefulness of a categorization of values. J Sports Med Phys Fitness. 2006;46:611–6. 115. Angeli A, Minetto M, Dovio A, Paccotti P. The overtraining syndrome in athletes: a stress-related disorder. J Endocrinol Investig. 2004;27:603–12. 116. Uusitalo AL, Huttunen P, Hanin Y, Uusitalo AJ, Rusko HK. Hormonal responses to endurance training and overtraining in female athletes. Clin J Sport Med. 1998;8:178–86. D. H. St-Pierre and D. Richard 54 117. Meeusen R, et al. Hormonal responses in athletes: the use of a two bout exercise protocol to detect subtle differences in (over)training status. Eur J Appl Physiol. 2004;91:140–6. https://doi.org/10.1007/ s00421-003-0940-1. 118. Lehmann M, Foster C, Dickhuth HH, Gastmann U. Autonomic imbalance hypothesis and overtraining syndrome. Med Sci Sports Exerc. 1998;30:1140–5. 119. Lehmann MJ, et al. Training and overtraining: an overview and experimental results in endurance sports. J Sports Med Phys Fitness. 1997;37:7–17. 120. Mor A, Kayacan Y, Ipekoglu G, Arslanoglu E. Effect of carbohydrate-electrolyte consumption on insulin, cortisol hormones and blood glucose after high-intensity exercise. Arch Physiol Biochem. 2019;125(4):344–50. https://doi.org/10.1080/13813 455.2018.1465098. 121. Svendsen IS, Killer SC, Gleeson M. Influence of hydration status on changes in plasma cortisol, leukocytes, and antigen-stimulated cytokine production by whole blood culture following prolonged exercise. ISRN Nutr. 2014;2014:561401. https://doi. org/10.1155/2014/561401. 122. Dmitrasinovic G, et al. ACTH, cortisol and IL-6 levels in athletes following magnesium supplementa- 123. 124. 125. 126. tion. J Med Biochem. 2016;35:375–84. https://doi. org/10.1515/jomb-2016-0021. Burman MA, Hamilton KL, Gewirtz JC. Role of corticosterone in trace and delay conditioned fear-potentiated startle in rats. Behav Neurosci. 2010;124:294–9. https://doi.org/10.1037/ a0018911. Cooper MA, Huhman KL. Blocking corticotropin-­ releasing factor-2 receptors, but not corticotropin-­ releasing factor-1 receptors or glucocorticoid feedback, disrupts the development of conditioned defeat. Physiol Behav. 2010;101:527–32. https://doi. org/10.1016/j.physbeh.2010.08.003. Row BW, Dohanich GP. Post-training administration of corticotropin-releasing hormone (CRH) enhances retention of a spatial memory through a noradrenergic mechanism in male rats. Neurobiol Learn Mem. 2008;89:370–8. https://doi.org/10.1016/j. nlm.2007.10.008. Chen Y, et al. Correlated memory defects and hippocampal dendritic spine loss after acute stress involve corticotropin-releasing hormone signaling. Proc Natl Acad Sci U S A. 2010;107:13123–8. https://doi. org/10.1073/pnas.1003825107. 4 Impact of Chronic Training on Pituitary Hormone Secretion in Humans Johannes D. Veldhuis and Kohji Yoshida Introduction The impact of chronic training on pituitary function is best understood by a basic appraisal of the neuroendocrine physiology of any given individual axis and the more complex interactive pathophysiology among axes [1–12]. Interaxes interactions have received relatively little attention. Even evaluating a single neuroendocrine axis in its dynamic state is a complicated challenge, given combined feedforward and feedback activities among the key control loci within any given axis [13, 14]. For example, in the case of the growth hormone (GH) and insulin-like growth factor 1 (IGF-1) axis, hypothalamic GH-releasing hormone (GHRH) secreted by arcuate nuclei stimulates pituitary GH secretion acutely, whereas the somatostatinergic system originating in the paraventricular nuclei opposes GHRH action [15]. These two neuronal inputs are reciprocally interconnected by intrahypothalamic synapses and common impinging neuromodulator pathways [14]. In addition, secreted GH feeds back on brain GH receptors, stimulating soma- J. D. Veldhuis (*) Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA e-mail: veldhuis.johannes@mayo.edu K. Yoshida Department of Obstetrics and Gynecology, University of Occupational and Environmental Health, Kitakyushu, Japan tostatin secretion and possibly inhibiting GHRH release. Available GH secreted into the bloodstream triggers IGF-1 production in various target tissues, and circulating IGF-1 is capable of inhibiting pituitary GH secretion indirectly and directly (see Fig. 4.1). Such feedforward (GHRHs driving GH secretion) and feedback (GHs inhibiting its own secretion, IGF-1 s inhibiting GH secretion, and so forth) dynamic control mechanisms in principle can be modified by the effects of exercise at one or more levels within the axis. Moreover, multiple determinants modulate neuroendocrine responses to training, such as the body composition of the individual, concurrent stress and/or weight loss, gender, diet and energy balance, concomitant drug or hormone use, age, puberty, pregnancy, and/or lactational status [16–18]. Here, we will examine the neuroendocrine determinants of pituitary responses to exercise training, explore some of the confounding issues (e.g., species differences, varying modes of neurohormone secretion, within- and between-axis regulation, and so on), and explore the overall notion of neuroendocrine axes as feedback and feedforward control systems capable of within-­ axis as well as between-axes interactions. Finally, metabolic mechanisms, although likely multifactorial, will be examined briefly, and their clinical implications underscored. © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_4 55 J. D. Veldhuis and K. Yoshida 56 GHRHp SRIHp FSRIH FGHRH Hypothalamus GHRHs SRIHs Elim GHp Elim Pituitary FGH GHs Elim IGF-Ip FIGF-I Systemic Circulation IGF-Is Elim Fig. 4.1 EU eumenorrheic athletes; AM amenorrheic athletes; ANX anorexics. Amenorrheic athletes have endocrine profiles (i.e., decreased thyroid hormones) similar to anorexics with chronic energy deficiency. (Data taken from Refs. [43, 47]). ∗Eumenorrheic means are significantly different from amenorrheic and anorexic means (p < 0.05). Panel A = total T3 (triiodothyronine) and panel B = total T4 (thyroxine) ultiple Determinants of Pituitary M Responses to Exercise Training training are not necessarily identical [20, 21, 23–30]. Moreover, stress or acute exercise imposed in an untrained individual will elicit endocrine responses potentially distinct from those observed in a highly physically trained subject [3, 8, 9, 11, 31–40]. Thus, many studies are confounded in part by the nature of the prior or concomitant training regimen, its duration, and its intensity. Finally, extreme physical exertion, “overreaching,” often evokes neuroendocrine disturbances that are not typical of either short-term submaximal exertion or chronic training [5, 9, 41–43]. Among other determinants of neuroendocrine responses to exercise training is the acuteness vs. chronicity of the training or exercise stimulus [2, 5, 11, 12, 19–22]. In particular, numerous studies demonstrate that acute exercise induces a variety of short-term changes in multiple hypothalamo–pituitary axes, including the nearly immediate secretion of GH and adrenocorticotrophic hormone (ACTH), β-endorphin, and cortisol, whereas the results of chronic 4 Impact of Chronic Training on Pituitary Hormone Secretion in Humans Neuroendocrine axes are exquisitely sensitive to nutrient intake, body composition, and total (and percentage) body fat [44–51]. Recent s­ tudies of the GH axis document unequivocally that percentage body fat, and in particular visceral (intra-abdominal) fat accumulation [52], negatively influences pulsatile GH secretion by suppressing the mass of GH secreted per burst and shortening the half-life of GH in the circulation [44, 45, 53–56]. The reciprocal relationship between visceral fat mass and GH secretion is illustrated in Fig. 4.2. Impaired GH secretion and more rapid GH removal jointly serve to reduce 24-h pulsatile serum GH concentrations in otherwise healthy but relatively more (viscerally) obese individuals. In contrast, acute weight loss or nutrient deprivation potently stimulates GH secretion in the human (while suppressing it in the rat) by 3–10-fold, with augmentation in both men and women of GH secretory pulse amplitude and mass and, to a lesser degree, burst frequency [47, 57, 58]. Consequently, nutrition, body weight, and body composition are prime determinants of pituitary MEAN 24-Hr Serum GH Concentration (µg/L) 2 57 (GH) secretory activity, which likely condition responses to exercise [59]. In addition, in men, as well as more recently recognized in women, body mass index (relative obesity) is a negative correlate of LH pulse amplitude [49, 60] and of the serum testosterone concentration in middle-aged men [49]. Gender distinctions also strongly influence the secretory output of several neuroendocrine axes. Foremost, the gonadotropin-releasing hormone– luteinizing hormone (GnRH–LH) follicle-­ stimulating hormone (FSH)–sex steroid axes in men and women exhibit clarion differences, particularly at the level of the so-called positive feedback, which is mechanistically required to achieve a preovulatory LH surge in women [61]. The GH–IGF-1 axis is also strongly sexually dimorphic in the human (as well as in the rat, as reviewed earlier [15]). For example, in healthy premenopausal men and women, GH secretion differs quantitatively by way of a nearly twofold greater mean (24-h) serum GH concentration, higher plasma IGF-1 level, greater mass of GH r = -0.68 p < 0.001 female male 1 0 3 5 4 6 Ln (Intraabdominal Fat) (cm2) Fig. 4.2 Negative relationship between 24-h mean serum GH concentration and intra-abdominal (visceral) fat mass, as determined by computerized axial tomographic scanning of the abdomen, in a cohort of healthy middle-aged men and women. GH concentrations were determined by 20-min blood sampling for 24 h and subsequent assay by immunofluorometry. The solid circles denote male subjects, and the open circles females. The regression line shows a strongly negative relationship between the natural logarithm of intra-abdominal adiposity and daily GH secretory activity in both men and women. In multiple linear regression analyses, intra-abdominal fat mass accounted for the majority of the variability in integrated serum GH concentrations, exceeding that owing to age and gender in this population. (Redrawn with permission from Vahl et al. [56]) J. D. Veldhuis and K. Yoshida 58 secreted per burst, and a more disorderly pattern of GH release in women compared to men [62]. In addition, the individual negative impact of age, body mass index, or percentage body fat on GH secretion is 1.5–2-fold more evident in men than women [48]; the positive effect of physical conditioning (increased VO2max) on GH release is also more prominent in the male [48] (Fig. 4.3). Integrated Serum GH Concentration (ug/L x min) Integrated Serum GH Concentration (ug/L x min) a 8000 6000 4000 2000 0 15 20 30 25 35 40 Integrated Serum GH Concentration (ug/L x min) Integrated Serum GH Concentration (ug/L x min) Age (yrs) 8000 6000 4000 2000 0 0 10 30 20 40 50 8000 6000 4000 2000 0 15 20 6000 4000 2000 0 20 30 40 50 60 70 Men V o 2 Max Women 40 % Change Per SD 35 VO2 Peak (ml/kg/min) 60 +33 +17 20 0 30 8000 Percent Body Fat b 25 BMI Age BMI % Fat –10 –20 –19 –16 –23 –40 –42 –45 –60 Fig. 4.3 (a) Impact of gender on the effects of age, adiposity as measured by body mass index (BMI) or percentage body fat, and physical fitness as quantitated by maximal oxygen consumption (VO2max peak or max) on integrated (24-h) serum GH concentrations in normal men (filled circles, N = 12) and women (open circles, N = 32). Linear regression plots are given for each sex. The solid lines denote regression in men, and the interrupted lines depict women’s data. (b) Approximately twofold greater impact of age, BMI, percentage body fat, and VO2max on 240-h mean serum GH concentrations in men than women. Data are means ± SEM expressed as standardized regression coefficients for the regression lines in (a). The gender-specific standardized regression coefficient is the slope of the linear relationship (given as a percentage) adjusted per unit standard deviation (SD) of the male or female group as pertinent. (Redrawn with permission from Weltman et al. [48]) 4 Impact of Chronic Training on Pituitary Hormone Secretion in Humans The tissue responses to GH also may be sex-­ specific in part, since estrogen can antagonize GH-driven IGF-1 production by the liver [15]. Consequently, gender must be identified as a major determinant of neuroendocrine responses in the GH–IGF-1 axis. Exercise-stimulated GH secretion may be less gender-dependent [63]. A lesser gender difference is observed for the corticotropin-releasing hormone (CRH)–arginine vasopressin (AVP)/ACTH–cortisol axis, where in the female, relatively increased expression of the CRH gene and increased adrenal responsiveness to ACTH are proposed [64]. However, the orderliness of individual 24-h ACTH and cortisol release (approximate entropy) or their relative synchrony (crossentropy) in men and women is similar [65]. Another significant confounding influence on neuroendocrine axes is age. For example, in the case of the LH–testosterone axis in men, there is progressive deterioration of LH or testosterone’s individual orderliness of release over 24 h and of LH–testosterone coupling or synchrony, when assessed by either cross-correlation analysis (indicating diminished feedforward control) [66] or cross-approximate entropy (indicating decreased pattern synchrony within the reproductive axis’ feedback system) [67]. The regularity of GH or ACTH/cortisol release also deteriorates with age in men and women [65, 68]. In addition, in both men and women, there are marked quantitative decreases in overall GH axis secretory activity, with a progressive fall in plasma IGF-1 and daily GH secretion rates with aging, especially in men compared to women of premenopausal age [44, 45, 48, 54]. Concurrent drug and/or hormone use can also markedly modify several pituitary-target tissue axes. For example, prescribed or self-use of anabolic steroids will profoundly suppress LH and FSH release and reduce levels of endogenous sex steroids while potentially stimulating the GH–IGF-1 axis (if aromatizable androgens are employed) [13, 63, 69–71]. Likewise, the use of birth control pills in young women stimulates GH secretion significantly and may produce some alterations in body composition [72]. At puberty, when sex steroid hormone secretion changes 59 more dramatically [73, 74], the individual’s GH– IGF-1 and/or GnRH–LH axis may be uniquely susceptible to the impact of exercise training (at least prior to pubertal onset), resulting in a significant delay in sexual maturation and adolescence and possibly reduced predicted adult height [75] (see Chap. 17; First edition). We infer that an array of important factors, such as exercise intensity and duration, its acuteness vs. chronicity, associated weight loss and/or stress (discussed further below), diet and energy balance, body composition, gender, age, and maturational status (e.g., prepubertal vs. pubertal), may all codetermine the neuroendocrine and pituitary responses to a stress perturbation, such as exercise. Other Confounding Issues One confounding issue experimentally in evaluating the impact of acute or chronic physical training on pituitary function is species differences. For example, in the rat, physical exertion reduces GH secretion [15], whereas in the human acute and chronic exercise, both increase GH secretion significantly, the former within 15–30 min and the latter following sustained exercise at an intensity above the individual lactate threshold [15, 20, 21, 24, 76, 77]. Indeed, chronic physical training in women results in a doubling of the 24-h mean serum GH level even on days when exercise is not undertaken [21] (see Fig. 4.4 [20]). Consequently, many experiments carried out in the rodent do not find applicability, especially for the GH–IGF-1 axis, to human studies. Moreover, the gender differences in the GH axis in the rat and human are readily distinguishable mechanistically in the two species, with a greater mean amplitude (and mass) of GH ­secretory bursts in women than men (but the converse occurs in the rat) [62]. A similarity in the two species is a more disorderly pattern of GH release in the female [78]. Further complicating interpretation and analysis of pituitary secretion are the multifold temporal modes of physiological pituitary hormone release: J. D. Veldhuis and K. Yoshida 60 SERUM GROWTH HORMONE CONCENTRATION (ug/L) a BASELINE 30 Control 30 30 • LT 15 15 15 0 0 0 30 30 > LT 1 YEAR > LT • LT 15 15 15 0 08:00 30 Control 10:00 0 08:00 08:00 10:00 08:00 0 08:00 10:00 08:00 10:00 08:00 TIME (CLOCKTIME) b BASELINE GROWTH HORMONE SECRETORY RATE (ug/L/min) 2 Control 2 2 • LT 1 1 1 0 0 0 2 2 > LT 1 YEAR > LT • LT 1 1 0 08:00 2 Control 10:00 1 0 08:00 08:00 10:00 08:00 0 08:00 TIME (CLOCKTIME) Fig. 4.4 The 24-hour serum GH concentration (a) or secretion rate (b) profiles in three different premenopausal women each studied twice: control (left; no exercise training, sedentary volunteer); before (baseline) and after 1 year of exercise training below or at the individually determined lactate threshold (LT) (middle panel; exercise volunteer #1), and exercise training above the LT (right panel; exercise volunteer #2). (Adapted with permission from Ref. [20]) 4 Impact of Chronic Training on Pituitary Hormone Secretion in Humans 1. Pulsatile. 2. Nyctohemeral or circadian. 3. Entropic, or moment-to-moment variations in the orderliness of secretion [67, 79–81]. Cell In contrast to the foregoing episodic (pulsatile) secretory mode are less rapid, 24-h variations in serum hormone concentrations, which are well established for ACTH, LH, GH, thyroid-­ stimulating hormone (TSH), prolactin, cortisol, and so forth [82]. These nyctohemeral (night– day) variations constitute only a small part of the total variation in daily neurohormone release. True circadian rhythms are so-called free-­running with a periodicity of 24 h, temperature-­ compensated, and susceptible to zeitgebers or specific phase-entraining cues [83]. Not all human 24-h neuroendocrine rhythms conform to this definition, which would denote true Secretory Velocity Pulsatile hormone secretion typically mirrors episodic neural input that acts via intermittent secretagog delivery to a responsive pituitary cell population in the absence of significant inhibitory input concurrently. Indeed, a pulse of pituitary hormone secretion can be viewed as a collection of secretory rates, centered about some moment in time. This concept is illustrated in Fig. 4.5. V1 V2 V3 Vi Vmax (Amplitude) Half-duration Burst Position Capillary Time II Concentration * Time Concentration Secretion 61 Time Time t S(z) Secretion Function * * E(t-z) = S(z)E(t-z)dz 0 Clearance Function Fig. 4.5 Schematized illustration of a model-specific deconvolution concept implemented to quantitate (GH) secretion. The upper landscape depicts an intuitive formulation of a hormone secretory burst, as arising from (multi-)cellular discharge of individual hormone molecules more or less in concert temporally, each at its own particular secretory rate (velocity). A secretory burst (or pulse) is visualized as an array of such molecular secretory velocities centered about some moment in time and dispersed around this center with a finite standard duration (SD) or half-width. The burst event may or may not be symmetric over time. The lower landscape with the algebraic subheads shows the mathematical notion, whereby a plasma hormone concentration peak (far right) = Convolution Integral is viewed as developing from a burst-like secretory process (far left) and a finite hormone-specific removal rate (half-life of elimination). The so-called convolution (intertwining or interaction) of the simultaneous secretory and elimination functions creates a resultant (skewed) plasma concentration pulse. Deconvolution analysis consists of mathematically estimating the constituent underlying secretory features (and/or associated half-life), given (a series of) blood hormone concentration peaks as the starting point. A variety of model-independent (waveform-­invariant) deconvolution strategies can also be applied, if a priori knowledge of the pertinent (biexponential) hormone elimination rate process is available. (Adapted with permission from Ref. [125]) 62 (suprachiasmatic nucleus-driven) circadian activity. Based on sleep-reversal studies, and so forth, circadian rhythmicity clearly does exist for ACTH/cortisol release in the human and GH secretion (approx 50% of the 24-h GH rhythm is sleep- and activity-entrained, and 50% is circadian) [15, 84]. Neurohormone release also exhibits features of minute-to-minute patterning, serial orderliness, or relative regularity, which can be quantified by an approximate entropy statistic [67, 78]. Higher values of approximate entropy denote greater disorderliness of hormone release and are a feature of female GH secretory patterns (compared to male), healthy aging of the human insulin, GH, LH, and ACTH/cortisol axes [54, 65, 67, 78, 85, 86], as well as aldosteronomas [87], tumoral pituitary hormone secretion (acromegaly, Cushing’s disease, and prolactinomas [65, 88]), and insulin release in type II diabetes mellitus [89, 90]. Thus, entropy measures can identify secretory disturbances complementary to pulsatile or circadian variations. The complex mode of pituitary hormone secretion imposes the need for appropriately rigorous sampling intensity and duration to capture the pulsatile, circadian, and entropic features, followed by application of relevant analytical tools appropriately validated under those conditions of study. Such technical issues have been reviewed recently [80, 91–93]. Further confounding in the literature arises because biochemically measurable endocrine changes do not always imply definite biological or clinical sequelae. For example, studies of the thyrotropin-releasing factor (TRH)–TSH–thyroidal axis have revealed numerous biochemically measurable changes during acute or chronic exercise, but their clinical sequelae are not known [94]. Similarly, in relation to the male reproductive axis, a variety of pituitary–gonadal changes are well established in response to chronic exercise, such as diminished LH pulse frequency at least in a subset of men, and relatively decreased spermatogenesis (e.g., a 30–50% decline in sperm number). However, clinical signs and symptoms of androgen deficiency rarely, if ever, occur, and male infertility is not known to be associated with J. D. Veldhuis and K. Yoshida chronic physical training [5, 32–35, 43, 95–100]. Finally, multiple hormones are produced by the anterior pituitary gland, and, as discussed further below, the corresponding individual axes may evince significant interactions. euroendocrine Axes as Feedback N and Feedforward Control Systems As intimated in the Introduction, neuroendocrine axes should be viewed as dynamic feedforward and feedback control systems. The term feedforward defines the ability of a secreted agonist to act on a remote or proximal tissue and evoke a typically sigmoidal (e.g., log-logistic) dose– response curve, e.g., as anticipated for GHRHs acting on somatotrope cells in the anterior pituitary gland, GnRHs acting on gonadotrope cells, and so forth [15, 101]. Conversely, feedback denotes the ability of a secreted product from a target tissue to inhibit the production of the agonistic signal, e.g., testosterone feeds back on hypothalamic GnRH secretion in the male, IGF-1 feeds back on pituitary somatotrope secretion of GH, l-thyroxine feeds back on TSH secretion at the pituitary and hypothalamic levels, and so forth. As highlighted in Figs. 4.1 and 4.6, both the GHRH–somatostatin/GH–IGF-1 axis [14] and the GnRH–LH/FSH/sex steroid [101] axes should be viewed as complex feedback and feedforward control systems [13, 14, 79, 101–103]. This concept is physiologically critical, since most pathophysiological stimuli impinge on several points within the feedback control system, thus impacting on the overall dynamics. Such system-level responses cannot be observed readily when separated components are studied individually. Similarly, the stress-responsive ACTH–adrenal axis comprises CRH–AVP/ ACTH–cortisol, with corresponding feedforward and interactive feedback mechanisms inherent [3, 40, 104]. An important notion in future studies of chronic exercise effects on the pituitary will be to limit isolation of individual components of the axis and rather study the overall axis dynamics. Technology, such as approximate entropy 4 Impact of Chronic Training on Pituitary Hormone Secretion in Humans (-) 63 Hypothalamus (-) H6 (-) H4 and H5 GnRH neurons GnRH Te Testis: Laydig Cell Time (+) Te H1 (basal Te) Pituitary Gland (-) H3 LH Time (+) H2 Time Fig. 4.6 Schematic illustration of the time-delayed negative feedback (−) and positive feedforward (+) within the human male GnRH–LH–testosterone (Te) axis. The broad arrows indicate feedforward (+) stimulus-secretion linkages, and the narrow arrows denote feedback (−) inhibi- tion. The “H” functions are developed further in Ref. [101] and serve to define the dose–response relationships at each feedback interface within the axis. (Adapted with permission from Ref. [101]) [67, 105] and network analysis [14, 101], for accomplishing the latter is just beginning to emerge. To date, the vast majority of published literature (as discussed throughout this volume) has enunciated changes at individual control points, which unfortunately subdivides the feedback system artificially and limits insights into its interactive properties, which function from minute to minute and day to day. experimental animals, alterations occur not only in hypothalamic GHRH and somatostatin gene expression but also in the GnRH neuronal ensemble and neuropeptide Y (NPY)- and CRH-­ secreting neurons [104, 106]. In conjunction with concurrent changes in dietary intake, activity of TRH neurons in the hypothalamus may also be suppressed (reviewed in Ref. [107]). Relevantly, these multiple neuronal pathways are directed by corresponding families of neurotransmitters (e.g., norepinephrine, serotonin, acetylcholine, and so forth), as well as various potent neuromodulators (e.g., NPY, galanin, and so on). Thus, a major focus in understanding the whole-body neuroendocrine responses of an intact organism to chronic exercise training must eventually include the articulation of not only individual neuronal pathway changes but also their collective and interconnected alterations owing to common neuromodulatory Introductions Among Neuroendocrine Axes Foremost among the challenges to be addressed in investigative and clinical neuroendocrine pathophysiology are the nature and mechanisms of interaction between two, or among three or more, neuroendocrine axes. For example, in relation to chronic exercise or other stressors in J. D. Veldhuis and K. Yoshida 64 Metabolic Mechanisms inputs. For example, infusion of leptin, the product of the ob gene in adipocytes, is capable of rescuing suppressed hypothalamic TRH secretion in fasting; relieving inhibited GnRH gene expression in certain stress models; and stimulating GH secretion in the fasted male rat (presumptively by reducing hypothalamic somatostatin gene expression). Thereby, leptin may integrate a complex response pattern via concerted hypothalamic actions that supervise diverse pituitary hormone secretory activities [107–109]. However, in the human, leptin levels correlate inversely (rather than directly, as in the rat) with GH axis secretory activity, as illustrated in Fig. 4.7 [55]. a Older Women Mean GH Concentration (µg/L) 3 P = 0.0072 r = -0.662 + + 2 + + 1 + + + 0 + + ++ + + 5 0 + 10 + 20 15 Leptin (ng/mL) b Women (fed/fasting) 10 Serum GH Concentration (µg/L) Fig. 4.7 (a) Inverse log-linear relationship between fasting serum leptin concentrations and integrated 24-h serum GH concentrations in 15 healthy postmenopausal women [55]. (b) Similar inverse (exponential) regression between serum leptin and GH output in young women fed or fasted [58]. P and r values for the linear regressions are shown. Adapted with permission The exact metabolic mechanisms that subserve hypothalamo–pituitary responses to exercise training are not known. Among those extensively considered are free fatty acids, which clearly can inhibit GH secretion [15]. On the other hand, any direct role of free fatty acids in modifying the GnRH–LH–gonadal axis is not evident. Similarly, both insulin and free IGF-1 can inhibit GH secretion directly at the anterior pituitary level and indirectly via hypothalamic effects under several conditions in certain species [15]. Moreover, prolonged nutrient and/or glucose deprivation can arrest puberty in the immature R = -0.683 P = 0.014 8 6 4 2 0 0 6 12 18 Serum Leptin Concentration (µg/L) 24 30 4 Impact of Chronic Training on Pituitary Hormone Secretion in Humans sheep and modify hypothalamic peptide secretion (e.g., stimulate CRH and/or AVP, while inhibiting GnRH, secretion) [104]. In contrast, carbohydrate ingestion during exercise in one study in the human seemed to increase cortisol and decrease gonadotropin release [110], whereas maintenance of euglycemia in another study abolished exercise-induced ACTH–cortisol release in nearly exhaustively exercised volunteers [111]. Finally, as intimated above, the peptide leptin can modify somatostatin, GnRH, TRH, and NPY gene expression, among other hypothalamic responses to the stress of fasting [55, 58]. Overall, we postulate that such multifactorial metabolic cues and the sex steroid milieu significantly codetermine neuroendocrine responses to exercise training [112–114]. In addition, under the most severe exercise stimulus, overall “finalcommon-pathway” stressor responses may prevail, such as secretion of reproductively inhibitory CRH and endogenous opioids, with consequent suppression of GnRH–LH secretion and conversely (in a species-specific manner) stressdriven alterations in the GH–IGF-­1 axis [10, 15, 38, 115–123]. Implications Among other implications of chronic training are favorable nonendocrine adaptations of hemodynamic and cardiovascular function. These changes are likely to be important in long-term health risk. Moreover, body compositional changes, motivated in part by the above neuroendocrine alterations, would be predicted to have a propitious impact on population-wide morbidity and mortality [12, 117]. In contrast, alterations in bone density accompanying chronic exercise have bipotential implications, e.g., with putatively increased fracture risk owing to sex steroid deprivation (amenorrhea) and possibly reduced total (height) growth potential [75] and, conversely, variably decreased fracture risk owing to increased bone density associated with the stress– strain mechanism of enhanced bone apposition accompanying sustained physical training [22, 124–126]. However, other confounding factors, 65 such as concurrent estrogen status, activity of the GH–IGF-1 axis, ethnicity, and gender, can also modify bone density and fracture risk. For example, we recently observed that black men and women show increased bone mass over their Caucasian counterparts but that only in men is the higher bone density in blacks associated with correspondingly increased GH secretion [127]. The mechanisms underlying such ethnic differences are also not yet understood, nor are possible ethnic differences in endocrine responsiveness to exercise stress well investigated. Summary The impact of chronic exercise training on the neuroendocrine control of the anterior pituitary gland, and its feedback and feedforward inputs, is complex. Multiple determinants influence adaptive hypothalamo–pituitary secretory responses to physical stress, namely, training intensity and duration, including overreaching exercise, concurrent weight loss, diet and energy balance, other associated stressors (both psychological and physical), body composition, gender, age, the sex steroid milieu, and developmental/maturational status. Confounding variables include interspecies differences, the complexity of neurohormone secretion (pulsatile, circadian, and entropic rhythms), the difficulty in interpreting earlier cross-sectional studies (with possible ascertainment bias) compared to longitudinal data, and the distinction between biochemical changes in and clinically significant sequelae of neurohormonal alterations with exercise. We emphasize that measurable pituitary responses to exercise should be viewed as part of a feedforward and feedback control system, as exemplified for the GH–IGF-1, GnRH–LH, CRH–AVP– ACTH, and other axes, with yet additional between-axes interactions. Although the final metabolic mechanisms that direct neuroendocrine changes in chronic training are not known definitively (e.g., free fatty acids, insulin, IGF-1, glucose, sex hormones, leptin, and/or others), their nature is likely multifactorial. In response to extremely strenuous exercise, stress-like neuro- J. D. Veldhuis and K. Yoshida 66 endocrine reactivity may predominate, whereas with appropriately modulated exercise intensity and volume, favorable clinical benefits, such as augmented GH secretion, cardiovascular conditioning, improved sense of well-being, and ­preserved reproductive function and bone density, likely ensue. Acknowledgments We thank Patsy Craig for her skillful preparation of the manuscript and Paula P. Azimi for the data analysis, management, and graphics. This work was supported in part by NIH Grant MO1 RR00847 (to the General Clinical Research Center of the University of Virginia Health Sciences Center), Research Career Development Award 1-KO4-HD-00634 (to J. D. V.), the Baxter Healthcare Corporation (Round Lake, IL, to J. D. V.), the NIH-supported Clinfo Data Reduction Systems, the University of Virginia Pratt Foundation and Academic Enhancement Program, the National Science Foundation Center for Biological Timing (Grant DIR89-20162), and the NIH NICHD U54 Center for Reproduction Research (HD96008). 9. 10. 11. 12. 13. 14. References 1. Veldhuis JD, Yoshida K, Iranmanesh A. The effect of mental and metabolic stress on the female reproductive system and female reproductive hormones. In: Hubbard J, Workman EA, editors. Handbook of stress medicine: an organ system approach. Boca Raton: CRC; 1997. p. 115–40. 2. Veldhuis JD, Evans WS, Weltman AL, Weltman JY, Rogol AD. Impact of exercise, as a paradigm of a physical stressor, on the female hypothalamo-­ pituitary-­gonadal axis. In: Genazzani AR, Pertraglia F, editors. Hormones in gynecological endocrinology. UK: Parthenon Publishing; 1992. p. 337–49. 3. Luger A, Deuster PA, Gold PW, Loriaux DL, Chrousos GP. Hormonal responses to the stress of exercise. Adv Exp Med Biol. 1988;245:273–80. 4. Bullen BA, Skrinar GS, Beitins IZ, Carr DB, Reppert SM, Dotson CO, et al. Endurance training effects of plasma hormonal responsiveness and sex hormone excretion. J Appl Physiol. 1984;56:1453–63. 5. Roberts AC, McClure RD, Weiner RI, Brooks GA. Overtraining affects male reproductive status. Fertil Steril. 1993;60:686–92. 6. Crist DM, Hill JM. Diet and insulin-like growth factor I in relation to body composition in women with exercise-induced hypothalamic amenorrhea. J Am Coll Nutr. 1990;9:200–4. 7. Loucks AB. Effects of exercise training on the menstrual cycle: existence and mechanisms. Med Sci Sports Exerc. 1990;22:275–80. 8. Wittet GA, Livesey JH, Espiner EA, Donald RA. Adaptation of the hypothalamopituitary adrenal 15. 16. 17. 18. 19. 20. 21. 22. axis to chronic exercise stress in humans. Med Sci Sports Exerc. 1996;28:1015–9. Viru A, Karelson K, Smirnova T. Stability and variability in hormonal responses to prolonged exercise. Int J Sports Med. 1992;13:230–5. Dishman RK. Brain monoamines, exercise, and behavioral stress: animal models. Med Sci Sports Exerc. 1997;29:63–74. Deuster PA, Chrousos GP, Luger A, DeBolt JE, Bernier LL, Trostmann UH, Kyle SB, Montgomery LC, Loriaux DL. Hormonal and metabolic responses of untrained, moderately trained, and highly trained men to three exercise intensities. Metabolism. 1989 Feb;38(2):141–8. Rogol AD, Weltman JY, Evans WS, Veldhuis JD, Weltman AL. Long-term endurance training alters the hypothalamic-pituitary axes for gonadotropins and growth hormone. In: Veldhuis JD, editor. Endocrinology and metabolism clinics of North America. Philadelphia: WB Saunders; 1992. p. 817–32. Veldhuis JD. Male hypothalamic-pituitary-gonadal axis. In: Yen SC, Jaffe RB, Barbieri RL, editors. Reproductive endocrinology. Philadelphia: WB Saunders; 1999. p. 622–31. Straume M, Chen L, Johnson ML, Veldhuis JD. Systems-level analysis of physiological regulation interactions controlling complex secretory dynamics of growth hormone axis: a connectionist network model. Methods Neurosci. 1995;28:270–310. Giustina A, Veldhuis JD. Pathophysiology of the neuroregulation of GH secretion in experimental animals and the human. Endocr Rev. 1998;19:717–97. Clapp JF. The effects of maternal exercise on early pregnancy outcome. Am J Obstet Gynecol. 1989;161:1453–7. Altemus M, Deuster PA, Galliven E, Carter CS, Gold PW. Suppression of hypothalamic–pituitary–adrenal axis responses to stress in lactating women. J Clin Endocrinol Metab. 1995;80:2954–9. Bonen A, Campagna P, Gilchrist L, Young DC, Beresford P. Substrate and endocrine responses during exercise at selected stages of pregnancy. J Appl Physiol. 1992;73:134–42. Veldhuis JD, Evans WS, Demers LM, Thorner MO, Wakat D, Rogol AD. Altered neuroendocrine regulation of gonadotropin secretion in women distance runners. J Clin Endocrinol Metab. 1985;61:557–63. Weltman A, Weltman JY, Schurrer R, Evans WS, Veldhuis JD, Rogol AD. Endurance training amplifies the pulsatile release of growth hormone: effects of training intensity. J Appl Physiol. 1992;76(6):2188–96. Rogol AD, Weltman A, Weltman JY, Seip RL, Snead DB, Levine S, et al. Durability of the reproductive axis in eumenorrheic women during one year of endurance training. J Appl Physiol. 1992;72(4):1571–80. Snead DB, Weltman JY, Weltman A, Evans WS, Veldhuis JD, Varma MM, et al. Reproductive hor- 4 Impact of Chronic Training on Pituitary Hormone Secretion in Humans 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. mones and bone mineral density in women runners. J Appl Physiol. 1992;72(6):2149–56. Alexander SL, Irvine CH, Ellis MJ, Donald RA. The effect of acute exercise on the secretion of corticotropin-releasing factor, arginine vasopressin, and adrenocorticotropin as measured in pituitary venous blood from the horse. Endocrinology. 1991;128:65–72. Thompson DL, Weltman JY, Rogol AD, Metzger D, Veldhuis JD, Weltman A. Cholinergic and opioid involvement in release of growth hormone during exercise and recovery. J Appl Physiol. 1993;75:870–8. Fryburg DA, Weltman A, Jahn LA, Weltman JY, Samolijik E, Veldhuis JD. Short-term modulation of the androgen milieu alters pulsatile but not exercise or GHRH-stimulated GH secretion in healthy men. J Clin Endocrinol Metab. 1997;82:3710–9. Wideman L, Weltman JY, Shah N, Story S, Veldhuis JD, Weltman A. The effects of gender on exercise-­ induced growth hormone (GH) release. J Applied Physiol. 1999;87:115–62. Kanaley JA, Weltman JY, Veldhuis JD, Rogol AD, Harman ML, Weltman A. Human growth hormone response to repeated bouts of aerobic exercise. J Appl Physiol. 1997;83:1756–61. Weltman JY, Seip RL, Weltman AL, Snead D, Veldhuis JD, Rogol AD. Release of luteinizing hormone and growth hormone after recovery from maximal exercise. J Appl Physiol. 1990;69(1):196–200. Pritzlaff CJ, Wideman L, Weltman JY, Abbott RD, Gutgesell ME, Hartman ML, Veldhuis JD, Weltman A. Impact of acute exercise intensity on pulsatile growth hormone release in men. J Appl Physiol. 1999 Aug;87(2):498–504. Weltman A, Pritzlaff CJ, Wideman L, Considine RV, Fryburg DA, Gutgesell ME, Hartman ML, Veldhuis JD. Intensity of acute exercise does not affect serum leptin concentrations in young men. Med Sci Sports Exerc. 2000 Sep;32(9):1556–61. Lehmann M, Knizia K, Gastmann U, Petersen KG, Khalaf AN, Bauer S, et al. Influence of 6-week, 6 days per week, training on pituitary function in recreational athletes. Br J Sports Med. 1993;27:186–92. Elias AN, Wilson AF. Exercise and gonadal function. Hum Reprod. 1993;8:1747–61. Cumming DC, Wheeler GD, MCColl EM. The effects of exercise on reproductive function in men. Sports Med. 1989;7:1–17. McColl EM, Wheeler GD, Gomes P, Bhambhani Y, Cumming DC. The effects of acute exercise on pulsatile LH release in high-mileage male runners. Clin Endocrinol. 1989;31:617–21. Hackney AC. Endurance training and testosterone levels. Sports Med. 1989;8:117–27. Caston AL, Farrell PA, Deaver DR. Exercise training-­ induced changes in anterior pituitary gonadotrope of the female rat. J Appl Physiol. 1995;79:194–201. Blaney J, Sothmann M, Raff H, Hart B, Horn T. Impact of exercise training on plasma adrenocor- 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 67 ticotropin response to a well-learned vigilance task. Psychoneuroendocrinology. 1990;15:453–62. Watanabe T, Morimoto A, Sakata Y, Wada M, Murakami N. The effect of chronic exercise on the pituitary-adrenocortical response in conscious rats. J Physiol. 1991;439:691–9. Fellmann N, Bedu M, Boudet G, Mage M, Sagnol M, Pequignot JM, et al. Inter-relationships between pituitary-adrenal hormones and catecholamines during a 6-day Nordic ski race. Eur J Appl Physiol Occup Physiol. 1992;64:258–65. Laatikainen TJ. Corticotropin-releasing hormone and opioid peptides in reproduction and stress. Ann Med. 1991;23:489–96. Bonen A, Keizer HA. Pituitary, ovarian, and adrenal hormone responses to marathon running. Int J Sports Med. 1987;3:161–7. Pestell RG, Hurley DM, Vandongen R. Biochemical and hormonal changes during a 1000 km ultramarathon. Clin Exp Pharmacol Physiol. 1989;16:353–61. Friedl KE, Plymate SR, Bernhard WN, Mohr LC. Elevation of plasma estradiol in healthy men during a mountaineering expedition. Horm Metab Res. 1988;20:239–42. Iranmanesh A, Lizarralde G, Veldhuis JD. Age and relative adiposity are specific negative determinants of the frequency and amplitude of growth hormone (GH) secretory bursts and the half-life of endogenous GH in healthy men. J Clin Endocrinol Metab. 1991;73:1081–8. Veldhuis JD, Iranmanesh A, Ho KKY, Lizarralde G, Waters MJ, Johnson ML. Dual defects in pulsatile growth hormone secretion and clearance subserve the hyposomatotropism of obesity in man. J Clin Endocrinol Metab. 1991;72:51–9. Veldhuis JD, Iranmanesh A, Evans WS, Lizarralde G, Thorner MO, Vance ML. Amplitude suppression of the pulsatile mode of immunoradiometric LH release in fasting-induced hypoandrogenemia in normal men. J Clin Endocrinol Metab. 1993;76:587–93. Hartman ML, Veldhuis JD, Johnson ML, Lee MM, Alberti KG, Samojlik E, et al. Augmented growth hormone (GH) secretory burst frequency and amplitude mediate enhanced GH secretion during a two-­ day fast in normal men. J Clin Endocrinol Metab. 1992;74:757–65. Weltman A, Weltman JY, Hartman ML, Abbott RA, Rogol AD, Evans WS, et al. Relationship between age, percentage body fat, fitness and 24 hour growth hormone release in healthy young adults: effects of gender. J Clin Endocrinol Metab. 1994;78:543–8. Veldhuis JD, Urban RJ, Lizarralde G, Johnson ML, Iranmanesh A. Attenuation of luteinizing hormone secretory burst amplitude is a proximate basis for the hypoandrogenism of healthy aging in men. J Clin Endocrinol Metab. 1992;75:52–8. Bergendahl M, Vance ML, Iranmanesh A, Thorner MO, Veldhuis JD. Fasting as a metabolic stress paradigm selectively amplifies cortisol secretory burst mass and delays the time of maximal nyctohemeral corti- J. D. Veldhuis and K. Yoshida 68 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. sol concentrations in healthy men. J Clin Endocrinol Metab. 1996;81:692–9. Aloi JA, Bergendahl M, Iranmanesh A, Veldhuis JD. Pulsatile intravenous gonadotropin-releasing hormone administration averts fasting-induced hypogonadotropism and hypoandrogenemia in healthy, normal-weight men. J Clin Endocrinol Metab. 1997;82:1543–8. Veldhuis JD, Kulin HE, Warner BA, Santner SJ. Responsiveness of gonadotropin secretion to infusion of an opiate-receptor antagonist in hypogonadotropic individuals. J Clin Endocrinol Metab. 1982;55:649–53. Iranmanesh A, Grisso B, Veldhuis JD. Low basal and persistent pulsatile growth hormone secretion are revealed in normal and hyposomatotropic men studied with a new ultrasensitive chemiluminescence assay. J Clin Endocrinol Metab. 1994;78:526–35. Veldhuis JD, Liem AY, South S, Weltman A, Weltman J, Clemmons DA, et al. Differential impact of age, sex-steroid hormones, and obesity on basal versus pulsatile growth hormone secretion in men as assessed in an ultrasensitive chemiluminescence assay. J Clin Endocrinol Metab. 1995;80:3209–22. Roubenoff R, Rall LC, Veldhuis JD, Kehayias JJ, Rosen C, Nicolson M, et al. The relationship between growth hormone kinetics and sarcopenia in postmenopausal women: the role of fat mass and leptin. J Clin Endocrinol Metab. 1998;83:1502–6. Vahl N, Jorgensen JOL, Skjaerback C, Veldhuis JD, Orskov HJ, Christiansen J. Abdominal adiposity rather than age and sex predicts the mass and patterned regularity of growth hormone secretion in mid-­ life healthy adults. Am J Phys. 1997;272:E1108–16. Ho KY, Veldhuis JD, Johnson ML, Furlanetto R, Evans WS, Alberti KG, et al. Fasting enhances growth hormone secretion and amplifies the complex rhythms of growth hormone secretion in man. J Clin Invest. 1988;81:968–75. Bergendahl M, Iranmanesh A, Evans WS, Veldhuis JD. Short-term fasting selectively suppresses leptin pulse mass and 24-hour rhythmic leptin release in healthy mid-luteal phase women without disturbing leptin pulse frequency or its entropy control (pattern orderliness). J Clin Endocrinol Metab. 1999;83:883–94. Walberg-Rankin J, Gwazdauskas FC. Response of beta-endorphin and estradiol to resistance exercise in female during energy balance and energy restriction. Int J Sports Med. 1992;13:542–7. Garcia-Rudaz MC, Ropelato MG, Escobar ME, Veldhuis JD, Barontini M. Augmented frequency and mass of LH discharged per burst are accompanied by marked disorderliness of LH secretion in adolescents with polycystic ovary syndrome. Eur J Endocrinol. 1998;139:621–30. Veldhuis JD, Johnson ML, Bolton WK. Analyzing pulsatile endocrine data in patients with chronic renal failure: a brief review of deconvolution techniques. Pediatr Nephrol. 1991;5:522–8. 62. Van den Berg G, Veldhuis JD, Frolich M, Roelfsema F. An amplitude-specific divergence in the pulsatile mode of GH secretion underlies the gender difference in mean GH concentrations in men and premenopausal women. J Clin Endocrinol Metab. 1996;81:2460–6. 63. Veldhuis JD, Metzger DL, Martha PM Jr, Mauras N, Kerrigan JR, Keenan B, et al. Estrogen and testosterone, but not a non-aromatizable androgen, direct network integration of the hypothalamo-somatotrope (growth hormone)-insulin-like growth factor I axis in the human: evidence from pubertal pathophysiology and sex-steroid hormone replacement. J Clin Endocrinol Metab. 1997;82:3414–20. 64. Roelfsema F, Van den Berg G, Frolich M, Veldhuis JD, van Eijk A, Buurman MM, et al. Sex-dependent alteration in cortisol response to endogenous adrenocorticotropin. J Clin Endocrinol Metab. 1993;77:234–40. 65. Van den Berg G, Pincus SM, Veldhuis JD, Frolich M, Roelfsema F. Greater disorderliness of ACTH and cortisol release accompanies pituitary-­dependent Cushing’s disease. Eur J Endocrinol. 1997;136:394–400. 66. Mulligan T, Iranmanesh A, Johnson ML, Straume M, Veldhuis JD. Aging alters feedforward and feedback linkages between LH and testosterone in healthy men. Am J Phys. 1997;42:R1407–13. 67. Pincus SM, Mulligan T, Iranmanesh A, Gheorghiu S, Godschalk M, Veldhuis JD. Older males secrete luteinizing hormone and testosterone more irregularly, and jointly more asynchronously, than younger males: dual novel facets. Proc Natl Acad Sci U S A. 1996;93:14100–5. 68. Keenan DM, Veldhuis JD, Sun W. A stochastic biomathematical model of the male reproductive hormone system. SIAM J Appl Math. 2000;61(3):934–65. 69. Rogol AD, Martha PM Jr, Johnson ML, Veldhuis JD, Blizzard RM. Growth hormone secretory dynamics during puberty. In: Adashi EY, Thorner MO, editors. The somatotrophic axis and the reproductive process in health and disease. New York: Springer; 1996. p. 69–82. 70. Veldhuis JD. Male hypothalamic–pituitary–gonadal axis. In: Lipshultz LI, Howards SS, editors. Infertility in the male. Philadelphia: Mosby-Year Book; 1996. p. 23–58. 71. Veldhuis JD, Iranmanesh A, Rogol AD, Urban RJ. Regulatory actions of testosterone on pulsatile growth hormone secretion in the human: studies using deconvolution analysis. In: Adashi EY, Thorner MO, editors. Somatotropic axis and the reproductive process in health and disease. New York: Springer; 1995. p. 40–57. 72. Veldhuis JD. Gender differences in secretory activity of the human somatotropic (GH) axis. Eur J Endocrinol. 1997;134:287–95. 73. Veldhuis JD. Neuroendocrine mechanisms mediating awakening of the gonadotropic axis in puberty. Pediatr Nephrol. 1996;10:304–17. 4 Impact of Chronic Training on Pituitary Hormone Secretion in Humans 74. Mauras N, Rogol AD, Haymond MW, Veldhuis JD. Sex steroids, growth hormone, IGF-I: ­neuroendocrine and metabolic regulation in puberty. Horm Res. 1996;45:74–80. 75. Theintz GE, Howald H, Weiss U, Sizonenko PC. Evidence for a reduction of growth potential in adolescent female gymnasts. J Pediatr. 1993;122:306–13. 76. Dawson-Hughes B, Stern D, Goldman J, Reichlin S. Regulation of growth hormone and somatomedinC secretion in postmenopausal women: effect of ­ physiological estrogen replacement therapy. J Clin Endocrinol Metab. 1986;63:424–32. 77. Weltman A, Seip RL, Snead D, Weltman JY, Evans WS, Veldhuis JD, et al. Exercise training at and above the lactate threshold in previously untrained women. Int J Sports Med. 1992;13(3):257–63. 78. Pincus SM, Gevers E, Robinson ICA, Roelfsema F, Hartman ML, Veldhuis JD. Females secrete growth hormone with more process irregularity than males in both human and rat. Am J Phys. 1996;270:E107–15. 79. Veldhuis JD. Pulsatile hormone release as a window into the brain’s control of the anterior pituitary gland in health and disease: implications and consequences of pulsatile luteinizing hormone secretion. Endocrinologist. 1995;5:454–69. 80. Veldhuis JD. Issues in quantifying pulsatile neurohormone release. In: Van de Kar LD, editor. Methods in neuroendocrinology: the cellular and molecular neuropharmacology series. Boca Raton: CRC; 1998. p. 181–203. 81. Veldhuis JD. New modalities for understanding dynamic regulation of the somatotropic (GH) axis: explication of gender differences in GH neuroregulation in the human. J Pediatr Endocrinol. 1996;9:237–53. 82. Veldhuis JD, Iranmanesh A, Johnson ML, Lizarralde G. Twenty-four hour rhythms in plasma concentrations of adenohypophyseal hormones are generated by distinct amplitude and/or frequency modulation of underlying pituitary secretory bursts. J Clin Endocrinol Metab. 1990;71:1616–23. 83. Veldhuis JD. A parsimonious model of amplitude and frequency modulation of episodic hormone secretory bursts as a mechanism for ultradian signaling by endocrine glands. In: Wever R, Kleitman N, editors. Ultradian rhythms in life processes: an inquiry into fundamental principles. London: Springer; 1992. p. 139–72. 84. Veldhuis JD, Iranmanesh A, Johnson ML, Lizarralde G. Amplitude, but not frequency, modulation of ACTH secretory bursts gives rise to the nyctohemeral rhythm of the corticotropic axis in man. J Clin Endocrinol Metab. 1990;71:452–63. 85. Hartman ML, Pincus SM, Johnson ML, Matthews DH, Faunt LM, Vance ML, et al. Enhanced basal and disorderly growth hormone (GH) secretion distinguish acromegalic from normal pulsatile GH release. J Clin Invest. 1994;94:1277–88. 69 86. Pincus SM, Veldhuis JD, Mulligan T, Iranmanesh A, Evans WS. Effects of age on the irregularity of LH and FSH serum concentrations in women and men. Am J Phys. 1997;273:E989–95. 87. Siragy HM, Vieweg WVR, Pincus SM, Veldhuis JD. Increased disorderliness and amplified basal and pulsatile aldosterone secretion in patients with primary aldosteronism. J Clin Endocrinol Metab. 1995;80:28–33. 88. Van den Berg G, Pincus SM, Frolich M, Veldhuis JD, Roelfsema F. Reduced disorderliness of growth hormone release in biochemically inactive acromegaly after pituitary surgery. Eur J Endocrinol. 1998;138:164–9. 89. Schmitz O, Porksen N, Nyholm B, Skjaerback C, Butler PC, Veldhuis JD, et al. Disorderly and nonstationary insulin secretion in glucose-tolerant relatives of patients with NIDDM. Am J Phys. 1997;35:E218–26. 90. Meneilly GS, Ryan AS, Veldhuis JD, Elahi D. Increased disorderliness of basal insulin release, attenuated insulin secretory burst mass, and reduced ultradian rhythmicity of insulin secretion in older individuals. J Clin Endocrinol Metab. 1997;82:4088–93. 91. Johnson ML, Veldhuis JD. Evolution of deconvolution analysis as a hormone pulse detection method. Methods Neurosci. 1995;28:1–24. 92. Veldhuis JD, Johnson ML. Specific methodological approaches to selected contemporary issues in deconvolution analysis of pulsatile neuroendocrine data. Methods Neurosci. 1995;28:25–92. 93. Veldhuis JD, Evans WS, Johnson ML. Complicating effects of highly correlated model variables on nonlinear least-squares estimates of unique parameter values and their statistical confidence intervals: estimating basal secretion and neurohormone half-­ life by deconvolution analysis. Methods Neurosci. 1995;28:130–8. 94. Loucks AB, Laughlin GA, Mortola JF, Girton L, Nelson JC, Yen SS. Hypothalamic–pituitary–thyroidal function in eumenorrheic and amenorrheic athletes. J Clin Endocrinol Metab. 1992;75:514–8. 95. Rogol AD, Veldhuis JD, Williams FT, Johnson ML. Pulsatile secretion of gonadotropins and pro-­ lactin in male marathon runners: relation to the endogenous opiate system. J Androl. 1983;5:21–7. 96. Bagatell CJ, Bremner WJ. Sperm counts and reproductive hormones in male marathoners and lean controls. Fertil Steril. 1990;53:688–92. 97. Arce JC, De Souza MJ. Exercise and male factor infertility. Sports Med. 1993;15:146–9. 98. Lucia A, Chicharro JL, Perez M, Serratosa L, Bandres EF, Legido JC. Reproductive function in male endurance athletes: sperm analysis and hormonal profiles. J Appl Physiol. 1996;81:2627–36. 99. Celani MF, Grandi M. The pituitary-testicular axis in non-professional soccer players. Exp Clin Endocrinol. 1989;94:244–52. J. D. Veldhuis and K. Yoshida 70 100. Vasankari TJ, Kujala UM, Taimela S, Huhtaniemi IT. Pituitary-gonadal response to gonadotropin-­ releasing hormone stimulation is enhanced in men after strenuous physical exercise. Acta Endocrinol. 1993;129:9–14. 101. Keenan DM, Veldhuis JD. A biomathematical model of time-delayed feedback in the human male hypothalamic–pituitary–Leydig cell axis. Am J Phys. 1998;275:E157–76. 102. Veldhuis JD, Johnson ML. Analytical methods for evaluating episodic secretory activity within neuroendocrine axes. Neurosci Biobehav Rev. 1994;18:605–12. 103. Keenan D, Veldhuis JD. A stochastic model of admixed basal and pulsatile hormone secretion as modulated by a deterministic oscillator. Am J Phys. 1997;273:R1182–92. 104. Bergendahl M, Veldhuis JD. Altered pulsatile gonadotropin signaling in nutritional deficiency in the male. Trends Endocrinol Metab. 1995;6:145–59. 105. Frager MS, Pieper DR, Tonetta SA, Duncan JA, Marshall JC. Pituitary gonadotropin-releasing hormone receptors: effects of castration, steroid replacement, and the role of gonadotropin-releasing hormone in modulating receptors in the rat. J Clin Invest. 1991;67(3):615–23. 106. Bergendahl M, Evans WS, Veldhuis JD. Current concepts on ultradian rhythms of luteinizing hormone secretion in the human. Hum Reprod Update. 1996;2:507–18. 107. Licinio J, Negrao AB, Mantzoros C, Kaklamani V, Wong M-L, Bongiorno PB, et al. Synchronicity of frequently-sampled 24-hour concentrations of circulating leptin, luteinizing hormone, and estradiol in healthy women. Proc Natl Acad Sci U S A. 1998;95:2541–6. 108. Schwartz MW, Seeley RJ, Campfield LA, Burn P, Baskin DG. Identification of targets on leptin action in rat hypothalamus. J Clin Invest. 1996;98:1101–6. 109. Ahima RS, Dushay J, Flier SN, Prabakaran D, Flier JS. Leptin accelerates the onset of puberty in normal female mice. J Clin Invest. 1997;99:391–5. 110. Vasankari TJ, Kujala UM, Viljanen TT, Huhtaniemi IT. Carbohydrate ingestion during prolonged running exercise results in an increase of serum cortisol and decrease of gonadotrophins. Acta Physiol Scand. 1991;141:373–7. 111. Tabata I, Ogita F, Miyachi M, Shibayama H. Effect of low blood glucose on plasma CRF, ACTH, and cortisol during prolonged physical exercise. J Appl Physiol. 1991;71:1807–12. 112. Matkovic V, Ilich JZ, Skugor M, Badenhop NE, Goel P, Clairmont A, et al. Leptin is inversely related to age at menarche in human females. J Clin Endocrinol Metab. 1997;82:3239–45. 113. Wade GN, Schneider JE, Li HY. Control of fertility by metabolic cues. Am J Phys. 1996;270:E1–19. 114. Mastrogiacomo I, Toderini D, Bonanni G, Bordin D. Gonadotropin decrease induced by prolonged exercise at about 55% of the VO2max in different 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. phases of the menstrual cycle. Int J Sports Med. 1990;11:198–203. Botticelli G, Bacchi Modena A, Bresciani D, Villa P, Aguzzoli L, Florio P, et al. Effect of naltrexone treatment on the treadmill exercise-induced hormone release in amenorrheic women. J Endocrinol Investig. 1992;15:839–47. Kanaley JA, Boileau RA, Bahr JM, Misner JE, Nelson RA. Cortisol levels during prolonged exercise: the influence of menstrual phase and menstrual status. Int J Sports Med. 1992;13:332–6. Hohtari H, Elovainio R, Salminen K, Laatikainen T. Plasma corticotropin-releasing hormone, corticotropin, and endorphins at rest and during exercise in eumenorrheic and amenorrheic athletes. Fertil Steril. 1988;50:233–8. Samuels MH, Sanborn CF, Hofeldt F, Robbins R. The role of endogenous opiates in athletic amenorrhea. Fertil Steril. 1991;565:507–12. Harber VJ, Sutton JR, MacDougall JD, Woolever CA, Bhavnani BR. Plasma concentrations of beta-­ endorphin in trained eumenorrheic and amenorrheic women. Fertil Steril. 1997;67:648–53. Hohtari H, Salminen-Lappalainen K, Laatikainen T. Response of plasma endorphins, corticotropin, cortisol, and luteinizing hormone in the corticotropin-­releasing hormone stimulation test in eumenorrheic and amenorrheic athletes. Fertil Steril. 1991;55:276–80. Armeanu MC, Lambalk CB, Berkhout GM, Schoemaker J. Effects of opioid antagonism with naltrexone on pulsatile luteinizing hormone secretion in women with hypothalamic amenorrhea in basal conditions and after discontinuation of treatment with pulsatile LHRH. Gynecol Endocrinol. 1992;6:3–12. Loucks AB, Mortola JF, Girton L, Yen SS. Alterations in the hypothalamic–pituitary– ovarian and the hypothalamic–pituitary–adrenal axes in athletic women. J Clin Endocrinol Metab. 1989;68:402–11. De Cree C, Van Kranenburg G, Geurten P, Fujimori Y, Keizer HA. 4-Hydroxycatecholestrogen metabolism responses to exercise and training: possible implications for menstrual cycle irregularities and breast cancer. Fertil Steril. 1997;67:505–16. Dugowson CE, Drinkwater BL, Clark JM. Nontraumatic femur fracture in an oligomenorrheic athlete. Med Sci Sports Exerc. 1991;23:1323–5. Baker E, Demers L. Menstrual status in female athletes: correlations with reproductive hormones and bone density. Obstet Gynecol. 1988;72:683–7. Marcus R, Cann C, Madvig P, Minkoff J, Goddard M, Bayer M, et al. Menstrual function and bone mass in elite women distance runners: endocrine and metabolic features. Ann Intern Med. 1985;102:158–63. Wright NM, Renault J, Willi S, Veldhuis JD, Gordon L, Key LL, et al. Greater secretion of growth hormone in black than in white males: possible factor in greater bone mineral density. J Clin Endocrinol Metab. 1995;80:2291–7. 5 Exercise and the GH-IGF-I Axis Alon Eliakim and Dan Nemet Introduction Physical activity and exercise play an important role in tissue anabolism, growth, and development, but the mechanisms that link patterns of physical activity with tissue anabolism are not completely understood. The anabolic effects of exercise are not limited to participants in competitive sports since substantial anabolic stimulus arises even from relatively modest physical activities [1]. The exercise-associated anabolic effects are age and maturity dependent. Naturally occurring levels of physical activity are significantly higher during childhood, and during adolescence there is a simultaneous substantial increase in muscle mass and strength. Thus, the combination of rapid growth, high levels of physical activity, and spontaneous pubertyrelated increases in anabolic hormones (growth hormone [GH], insulin-­ like growth factor-I [IGF-I], and sex steroids) suggests the possi- A. Eliakim (*) Endocrinology Clinic, Meir Medical Center, Sackler School of Medicine, Tel Aviv University, Department of Pediatrics, Kfar Saba, Israel e-mail: eliakim.alon@clalit.org.il D. Nemet Child Health and Sports Center, Meir Medical Center, Sackler School of Medicine, Tel Aviv University, Department of Pediatrics, Kfar Saba, Israel bility of integrated mechanisms relating exercise with anabolic responses. In contrast, participation of young athletes in intense competitive training, especially if associated with inadequate caloric intake, may be associated with health hazards and may reduce growth potential [2]. Training efficiency depends on the exercise intensity, volume, duration, and frequency and on the athlete’s ability to tolerate it. An imbalance between the training load and the individual’s tolerance may result in under- or overtraining. Therefore, efforts are made to develop objective methods to quantify the fine balance between training load and the athlete’s tolerance. The endocrine system, by modulation of anabolic and catabolic processes, seems to play an important role in the physiological adaptation to exercise training [3]. In recent years changes in circulating components of the GH-IGF-I axis, a system of growth mediators that control somatic and tissue growth [4], have been used to quantify the effects of training [5]. Interestingly, exercise is also associated with remarkable changes in catabolic hormones and inflammatory cytokines [i.e., interleukin-6 (IL-6)], and the exercise-related response of these markers can be also used to gauge exercise load [6, 7]. Anabolic response dominance will eventually lead to increased muscle mass and improved fitness, while prolonged dominance of the catabolic response, particularly if combined with inadequate nutrition, may © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_5 71 72 ultimately lead to overtraining. Therefore, the ­evaluation of changes in these antagonistic circulating mediators may assist in quantifying the effects of different types of single and prolonged exercise training and recovery modalities. This chapter demonstrates the effects of exercise on the GH-IGF-I axis, with an emphasis on the unique relationships between the exercise-­ related anabolic response and exercise-associated changes in inflammatory mediators. An important goal of this chapter is to show how exercise-­ induced changes in the GH-IGF-I-inflammatory axis can be used by elite athletes and their accompanying staff to evaluate training load throughout the competitive season and in the preparation for competition in “a real-life” setting. Finally, the chapter demonstrates new data on the possible use GH-IGF-I genetics in sports selection and prediction of excellence. A. Eliakim and D. Nemet paracrine secretion and regulation, which are only partially GH dependent. IGF-I is responsible for most, but not all, anabolic and growth-­ related effects of GH. IGF-I stimulates SMS secretion and inhibits GH by a negative feedback mechanism [9]. The bulk of circulating IGF-I is bound to IGFBPs. The most important circulating BP is IGFBP-3, which accounts for 80% of all IGF binding. Some IGFBPs are GH dependent (e.g., IGFBP-3), while others (IGFBP-1 and IGFBP-2) are insulin dependent (being high when insulin level is low). The interaction between IGF-I and its BPs is even more complicated since some BPs stimulate (e.g., IGFBP-5), while others inhibit (e.g., IGFBP-4) IGF-I anabolic effects [10]. Some hormones in the GH-IGF-I axis (i.e., GHRH and GH) have a pulsatile secretion pattern, and it has been shown that GH pulsatility is important for growth rate acceleration [11]. In contrast, IGF-I and IGFBPs level are relatively stable throughout the day. The GH-IGF-I Axis Furthermore, several components of the axis The GH-IGF-I axis is composed of hormones, are age and maturity dependent. GH, GHBP, growth factors, binding proteins (BP), and recep- IGF-I, and IGFBP-3 reach their peak levels durtors that regulate essential life processes. The ing puberty [12] and decrease with aging [13]. axis starts at the central nervous system where These changes are partially sex hormone mediseveral neurotransmitters (e.g., catecholamines, ated. Nutritional status influences the GH-IGF-I serotonin, cholinergic agents, etc.) stimulate the axis as well. Prolonged fasting and malnutrition hypothalamus to synthesize growth hormone-­ increase GH secretion, yet despite elevated GH, releasing hormone (GHRH) and somatostatin IGF-I levels remain low due to reduced levels of (SMS). GHRH stimulates the anterior pituitary to GH receptors [14]. All these factors must be taken into account when studying the effect of secrete GH, while SMS inhibits GH secretion. GH is the major product of the axis. One of exercise on the GH-IGF-I axis. GH most important functions is the stimulation of hepatic IGF-I synthesis. However, some GH effects on metabolism, body composition, and Optimizing Training Modalities tissue differentiation are IGF-I independent. Tissue GH bioactivity results from interaction Aerobic Training between GH and its receptor. The GH receptor is composed of intra- and extracellular transmem- The majority of the current knowledge regarding brane domains. The extracellular domain is iden- the importance of the GH response to exercise is tical in structure to GH-binding protein (GHBP) based on studies examining the effect of aerobic-­ [8]; thus, measuring circulating GHBP levels type exercise in individualized sports [15, 16]. To this end, when exercise is performed at the same reflects GH receptor number and activity. IGF-I is one of the insulin-related peptides. absolute intensity, the GH response is greater in Some of IGF-I effects are GH dependent, but the less fit subjects [17]. Yet, when subjects perform majority of its actions occur due to autocrine or exercise at the same absolute, rather than relative 5 Exercise and the GH-IGF-I Axis intensity, some individuals exercise below, while others exercise above, their lactic/anaerobic threshold (LAT). This is important since circulating GH levels increase only in response to aerobic exercise intensity above the LAT and because exercise loads of 75–90% of the maximal aerobic power yielded greater GH increase than milder loads. Therefore, results of studies in which the GH response to exercise was tested at an absolute work rate demonstrate simply that as individuals become fitter, the stress associated with exercise at an absolute work rate is reduced. The obvious implication for athletes is that as they become more physically fit, a more intense exercise should be performed to stimulate GH secretion. This is consistent with the common coaching modality of training cycles with workloads of increased intensity throughout the training season. The duration of aerobic exercise for the stimulation of GH secretion should be at least 10 min [18]. The exercise-induced GH peak occurs 25–30 min after the start of exercise (slightly earlier in females compared to males), irrespective to its duration [19, 20]. Thus, when the exercise task is brief (e.g., 10 min), GH peak is reached after the cessation of exercise, while, when exercise is long (e.g., 60 min), GH peak is reached while the individual is still exercising. The important possible implication for athletes is that brief training sessions can be enough to stimulate the GH-IGF-I axis and to achieve a “training effect” (i.e., relative to this hormone and its response). Pituitary refractoriness, a time in which the normal pituitary gland will not respond sufficiently to any stimulus for GH release, could also influence the GH response to exercise. For example, the GH response to exercise was inhibited if a spontaneous, early morning, GH pulse had occurred within 1 hour prior to the exercise test [20]. A refractory period of at least 1 hour was also shown following exercise-induced GH secretion (i.e., the subsequent GH response to exercise was attenuated) [21]. GH auto-­inhibition, exercise-induced elevation in free fatty acids, or alterations in parasympathetic-sympathetic tone can explain the development of pituitary refractoriness. A recovery from pituitary refractoriness to GH secretion was seen if a second bout of high-­ 73 intensity endurance exercise was performed 3 hours after the first session [22]. Consistent with this report, integrated 1.5 hours GH concentrations were significantly greater if differences between the exercise bouts (30 min, 70% VO2 max [maximal oxygen uptake]) were 3.5 hours and not when 1 hour apart [23]. The practical application for athletes should therefore be that in order to achieve optimal GH secretion, the rest interval between multiple daily training sessions should be long enough (probably more than 3 hours) to allow pituitary recovery. Anaerobic Exercise A major progress was achieved in recent years in the understanding of the effects of anaerobic exercise on the GH-IGF-I axis. Stokes et al. [24] studied the effect of a single supramaximal 30 sec sprint on a cycle ergometer against different levels of resistance workloads. They found that the increase in GH levels was significantly greater when resistance was 7% (faster cycling) and not 9% (slower cycling) of body mass. Consistent with that, it was shown that when heavier loads were lifted, more total work was performed, and higher IGF-I levels were found using faster compared to slower tempo resistance training [25]. The possible implication for athletes is that lower levels of resistance and/or faster anaerobic efforts may better stimulate the GH-IGF-I axis and thus preferred by coaches and athletes. Interval training is currently one of the most frequent training methods used in anaerobic and aerobic-type sports [26]. The intensity of such training depends on the running distance (sprint versus long distance), running speed (percent of maximal speed), the number of repetitions, and the length of the rest interval between the runs. In addition, coaches and athletes often change the style of the interval training and use constant running distances (e.g., 6 × 200m), increasing distance interval session (e.g., 100 m–200 m–300 m–400 m), decreasing distance interval session (e.g., 400 m–300 m–200 m–100 m), or a combination of increasing-decreasing distance interval session A. Eliakim and D. Nemet 74 (e.g., 100 m–200 m–300 m–200 m–100 m). While these style differences may seem negligible, they may involve different physiological demands, since in the increasing distance protocol, metabolic demands (e.g., lactate levels) increase gradually and are highest toward the end of the session, while in the decreasing distance protocol, the metabolic demands are higher from the beginning of the session [27], if the intensity of the intervals is appropriate and able to be maintained by the athlete. A significant increase in GH and IL-6 levels was demonstrated following a typical constant distance (4 × 250m) interval training [28]. Consistent with previous findings in aerobic exercise, changes in the GH-IGF-I axis following the brief sprint interval exercise suggested exercise-­ related anabolic adaptations. The increase in IL-6 probably indicates its important role in muscle tissue repair following anaerobic exercise [29]. It was suggested that changes in the anabolic/catabolic/inflammatory balance can be used as an objective tool to gauge the training intensity of different types of anaerobic exercises and training periods as well. More recently, we evaluated the effect of increasing (100–200–300–400 m) and decreasing distance (400–300–200–100 m) sprint interval training protocols, two other common types Decreasing 12 of sprint interval training, on the balance between anabolic, catabolic, and inflammatory mediators [27]. Both types of sprint interval trainings led to a significant increase in lactate and the anabolic factors GH and IGF-I. Both types of sprint interval sessions led to a significant increase in the circulating inflammatory mediators (IL-6). Interestingly, the lactate and GH area under the curve was significantly greater in the decreasing distance session. In contrast, rate of perceived exertion (RPE) was higher in the increasing distance session. Thus, despite similar running distance, running speed, and total resting period in the two interval training sessions, the decreasing distance interval was associated with a greater metabolic (lactate) and anabolic (GH) response (see Fig. 5.1). Interestingly, these greater metabolic and anabolic responses were not accompanied by an increase in RPE suggesting that physiological and psychological responses to interval training do not necessarily correlate. When the athletes were asked to explain why the increasing distance protocol was perceived as more intense, they replied that the fact that the longest and hardest run (400 m) was only at the end of the session was very difficult to tolerate. Coaches and athletes should be aware of these differences and, as a consequence, of the need for Increasing 40 10 30 8 6 20 4 10 2 0 Growth hormone (ng/ml) 0 Fig. 5.1 The effect of decreasing and increasing distance of sprint interval exercise on GH and GH area under the curve responses. The decreasing distance interval was Growth hormone AUC (ng\ml) associated with a greater anabolic [GH (left) and GH AUC (right)] response 5 Exercise and the GH-IGF-I Axis specific recovery adaptations after different types of interval training sessions. Differences in physiological and psychological responses to competitive sport training, and their influence on the training course and recovery process, should also be better addressed in future research work. Finally, in contrast to the observation that both aerobic and anaerobic exercise require a high metabolic demand in order to stimulate GH secretion, we previously demonstrated a small but significant GH response to an exercise input that was perceived as difficult by the participants (i.e., 10 min of unilateral wrist flexion, a small and relatively unused muscle group) but which had no effect on heart rate or circulating lactate levels [30, 31]. This suggests that factors like the individual’s perceived exertion and associated psychological stress play an important role in the activation of the hypothalamic-pituitary axis and to GH release even in exercise protocols involving small muscle groups. Resistance Exercise Previous studies have demonstrated increases in GH following a session of resistance exercise in adolescent and prepubertal boys. Children and adolescents demonstrate a lower GH response to resistance exercise compared with adults, presumably due to higher baseline GH levels [32]. As mentioned before, growth hormone is secreted in a pulsatile manner, with highest secretion during deep sleep, especially in children. Interestingly, Nindl et al. [33] reported that in men, an afternoon resistance training session affected the GH secretion pattern during resting states. Specifically, while mean GH secretion did not change, a lower rate of secretion in the first half of sleep and a higher rate of secretion in the second half of sleep were detected. This could be a direct effect of the resistance exercise on GH secretion, or an indirect effect on sleep quality. Considering the importance of nighttime GH secretion for linear growth, resistance training in adolescent athletes during different times of day (e.g., morn- 75 ing vs afternoon) may have different effects on sleep quality and/or directly on GH secretion pattern. Likewise, effects on the pulsatile secretion pattern may potentially even have effects on their linear growth. There is certainly a need for further research in this area. Team Sports Very few studies examined the effect of exercise on the GH-IGF-I axis in team sports. We previously demonstrated an increase in GH, testosterone, and IL-6 levels following a typical volleyball practice in adolescent national team level male and female players [34]. Interestingly, one of our most important findings was the effect of training on the endocrine response to a single practice. The hormonal response to a typical 60 min volleyball practice was assessed before and after 7 weeks of training during the initial phase of the season in elite national team level male and female players. In male players [35], training resulted in significantly greater GH increase along with significantly reduced IL-6 response to the same relative intensity volleyball practice. In female players [36], training resulted in significantly lower cortisol and IL-6 increase to the same relative intensity volleyball practice. The results suggest that along with the training-­ associated improvement of power, anaerobic and aerobic characteristics, part of the adaptation to training is that a single practice becomes more anabolic and less catabolic/inflammatory as training progresses during the initial phases of the training season (Fig. 5.2). Hormonal measurements therefore may assist athletes and their coaching staff in assessing the training program adaptation throughout different stages of the competitive season. Finally, higher social position was associated with higher levels of IGF-I in both men and women, independent of wide range of known confounders such as age, ethnicity, body weight, and nutrition [37]. Along this line, Bogin et al. [38] studied high-level male and female competitive athletes from different university team sports (men, lacrosse, handball, rugby, and volleyball; A. Eliakim and D. Nemet 76 5 Lactate nmol/L 4 3 2 1 0 2.5 Pre training IL-6 pg/ml 2.0 Post training 1.5 1.0 .5 0.0 Fig. 5.2 The effect of training on the hormonal response to a single volleyball practice in male adolescent players. Same level of training (i.e., lactate response) leads to a reduced inflammatory response (IL-6) Fig. 5.3 A proposed exercise-training-IGF-I cycle. With proper training, both single practice and prolonged training increase IGF-I levels, which in turn may increase the chances of an athlete to win women, football, rugby, netball, and volleyball) and assumed that what determines the social position in this social network is the level of ­success in sports (and not the economic status). Therefore the athletes were divided into winners and losers. The main finding of the study was that both pre- and post-competition IGF-I levels were about 11% greater among winners. There was no difference in the competition-related changes in IGF-I levels between the groups, suggesting that it is the baseline levels of IGF-I and not the change in IGF-I levels during the competition that may contribute to winning. This is the first study that related IGF-I levels with winning. It seems that IGF-I levels integrate the multiple genetic, nutritional, social, and emotional influences to a coherent signal that regulates growth and possibly athletic performance. This suggests a novel cycle: both single practice and prolonged training increase IGF-I levels, which in turn increase the chances of an athlete to win (see proposed model in Fig. 5.3). However, future larger studies that analyze other types of team sports and individual sports and that provide better control for nutritional, training, and doping status are needed to confirm this very interesting finding. Single practice IGF Polymorphism ul e f ss c uc S Prolonged training N on su cc es sf ul IGF-I IGF-I Winning Losing 5 Exercise and the GH-IGF-I Axis “Real-Life” Exercise Studies The majority of studies on the effect of exercise on the GH-IGF-I axis are laboratory-based. There is no doubt that laboratory-based science is important for understanding the exercise-related endocrine response. However, the translation of this knowledge to everyday use of competitive athletes is complicated, and there is a severe lack of “real-life” setting studies on the endocrine effect of exercise training. One of the main obstacles of executing “real-life” training studies is exercise standardization. We recently compared previous reports on the effect of “real-life” typical field individual (i.e., cross-country running and wrestling – representing combat versus noncombat sports) and team sports practice (i.e., volleyball and water polo – representing water and land team sports) on GH levels [39]. In this study, we were unable to control for the participants’ fitness level or for each practice’s intensity. In order to achieve some standardization, however, participants did not train during the day before the study, the duration of each practice was limited to 60–90 min, and the practice was performed during the initial phases of the training season when athletes are in relatively lower fitness level. All practice sessions were performed in the morning hours, and each typical practice included warm-up, main training segment, and cooldown. Blood samples were collected immediately before and at the end of practice, and the effect of the typical practice on hormonal and cytokine levels was expressed as percent change. Despite some limitations within the study, several important observations and conclusions could be drawn about the “real-life” training-­ related GH response from this unique comparison. These include the following: (1) cross-country running practice and volleyball practice in both males and females were associated with significant increases of GH, (2) the magnitude (percent change) of the GH response to the different ­practices was determined mainly by what were the pre-exercise GH levels, (3) there was no difference in the GH response 77 between individual and team sports practices, and (4) interestingly, the GH response to the typical practices was not influenced by the practiceassociated lactate change. Cryotherapy, Recovery, and the GH-IGF-I Axis The development of methods to enhance the recovery of elite athletes from intense training and/or competition has been a major target of athletes and their accompanying staff for many years. Cryotherapy is widely used to treat sports-­ associated traumatic injuries and as a recovery modality following training and competition that may cause some level of traumatic muscle injury [40, 41]. However, evidence regarding the effectiveness and appropriate guidelines for the use of cryotherapy are limited. To this end, Nemet et al. studied the effect of cold ice pack application following a brief sprint interval training on the balance between anabolic, catabolic, and circulating pro- and anti-inflammatory cytokines evaluated in 12 male, elite junior handball players [42]. The interval practice (4 × 250m) was associated with a significant increase in GH and IL-6 levels. Local cold-pack application was associated with significant decreases in the anabolic factors IGF-I and IGF-binding protein-3 during the recovery from exercise, supporting some clinical evidence of possible negative effects of cryotherapy on hormonal responses. These results, along with no clear detected effect on muscle damage or delayed onset muscle soreness (DOMS), may suggest that the use of cold packs should probably be reserved for traumatic injuries or used in combination with active recovery and not with complete rest. However, the findings of this study illustrate how exercise-induced changes in the GH-IGF-I axis and other catabolic and inflammatory markers may be used as an aid in competitive training. Further studies are needed to explore the beneficial use of anabolic, catabolic, and inflammatory markers measurement in the “monitoring” of recovery from exercise. 78 Nutrition, Performance, and the GH-IGF-I Axis Nutritional factors may intervene with the GH response to exercise. For example, intravenous administration of the amino acid arginine is a strong stimulator of GH release and therefore used, for example, as one of the more common provocation tests for GH secretion in the diagnosis of some clinical states (e.g., short stature). Recently, it was demonstrated that oral arginine stimulates GH secretion as well [43]. Therefore, it is possible that the ingestion of arginine prior to exercise may attenuate the exercise-related GH response, most probably due to induction of a refractory period [44]. Along these lines, ingestion of a lipid-rich meal 45–60 min prior to an intermittent 30 min cycle ergometer exercise resulted in a significant more than 40% reduction in the exercise-induced GH elevation in healthy children [45]. The effect of prior high-fat meal ingestion appeared to be GH selective, as other counter-regulatory hormone responses to exercise, such as glucagon, cortisol, and epinephrine, were not affected. Similarly, administration of high-fat meal attenuated the magnitude of GH response to exercise also in adults, and this inhibition was correlated with circulating levels of SMS [46]. Interestingly, a high carbohydrate meal with a similar caloric content was also associated with a small decrease in GH response to exercise; however, this decrease was not statistically significant. These studies indicate that food consumption prior to exercise or sporting practice should be carefully selected, since a consumption of high-fat meal may affect the hormonal response to a training session. Very few studies have examined the effects of nutrition on longer periods of training (i.e., weeks or months) primarily due to logistical issues. The timing of nutritional supplementation may also affect the training-associated response of the GH-IGF-I axis. For example, the combination of post-exercise essential amino acids and carbohydrate supplementation was accompanied by significant increases in free IGF-I during 3 weeks of high-intensity interval training [47] (compared to carbohy- A. Eliakim and D. Nemet drate only or placebo). Protein supplementation 1 hour before and after practice during 10 weeks of resistance training (four times/week) were more effective than carbohydrate placebo in increasing muscle mass and muscle strength and were associated with greater increases in IGF-I and IGF-I mRNA in untrained males [48]. Consistent with these findings, twice daily protein compared to carbohydrate supplementation during 6 months of strength and conditioning training (five times/week) was associated with greater increase in IGF-I levels in untrained late pubertal and young adult males and females [49]. These results as well as others suggest a beneficial effect for protein supplementation during prolonged period of resistance training, but more research is needed on this topic to provide specific amount and type of protein supplementation. Amenorrhea, Performance, and the GH-IGF-I Axis The inhibitory effect of exercise training, in particular, when associated with nutritional deprivation, on the pulsatile release of hypothalamic GnRH and pituitary LH and FSH secretion is well established and will be discussed in more detail in other chapters of this book (see Chaps. 4, 7, and 8). This inhibition results in increased risk of athletic amenorrhea and hypoestrogenism [50]. To this end, it was shown that the exercise-­ associated GH release is attenuated in amenorrheic athletes. The mechanism for the attenuated amenorrhea-associated exercise-induced GH response is not completely understood. However, it was found recently [51] that low estrogen leads to decreased post-exercise type 1 deiodinase (an enzyme that converts T4 to the more active thyroid hormone T3), reduced T3 levels, and in turn a blunted GH response. This is particularly relevant to the adolescent female athlete, since the prevalence of amenorrhea among these athletes is 4–20 times higher than the general population [52]. “Athletic amenorrhea” appears mainly in younger athletes and is associated with sports activities where leanness provides a competitive 5 Exercise and the GH-IGF-I Axis 79 advantage (e.g., aesthetic-type sports, long-­ distance running, etc.) and, in particular, where intense training is accompanied by inadequate nutrition [50].The reduced exercise-induced GH response in these athletes should be considered critically important since it indicates probably reduced training effectiveness and performance. Consistent with that Vanheest et al. [53] showed that reduced energy intake and availability that was associated with ovarian suppression was also accompanied by lower T3 and IGF-I levels and by a 9.8% decline in 400 m swim velocity compared to 8.2% improvement among female swimmers without ovarian suppression at the end of 12 weeks of training (N.B., in total, 18% difference!). This occurred despite similar training protocols and while the ovarian suppressed swimmers were still menstruating (although less regularly). This is important because some coaches and young athletes promote energy-­ restrictive practices with the belief that it improves competitive performance [54]. The results of this study emphasize that athletes can maintain chronic energy deficit for varied periods with continued success in sport; however, prolonged negative energy balance results in training maladaptation and reduced performance. This may be particularly relevant for athletes during adolescence, a time with greater energy needs for growth and maturation. Measurements of hormones and in particular IGF-I levels can also assist athletes and coaches in the training preparation for selected competitions. For example, in one study, the effect of 4 weeks of training on fitness, self-assessment physical conditioning scores, and circulating IGF-I were determined in elite professional handball players [55] during their preparation for the junior world championships. Training consisted of 2 weeks of intense training followed by 2 weeks of relative tapering. Circulating IGF-I and physical conditioning scores decreased initially and returned to baseline levels at the end of tapering. There was a significant positive correlation between the changes in circulating IGF-I and self-assessed physical conditioning scores suggesting that the player’s self-assessment may be a somewhat reliable tool when laboratory assessment is unavailable (see Fig. 5.4). Consistent with these findings, a follow-up of IGF-I levels during a training season in elite adolescent wrestlers showed an initial decrease in IGF-I level during periods of heavy training and return to baseline during tapering down and prior to the competition season [56]. Interestingly, changes in the pro-inflammatory mediators IL-6 correlated negatively with changes in IGF-I, being high when IGF-I level was low, and normalized ∆ Change in self assessment physical conditioning scores 50 ∆ Change in circulating IGF-I (ng/ml) Fig. 5.4 Relationship between changes in self-assessment physical conditioning scores and change in circulating IGF-I during 2 weeks of intense training in handball players. There was a significant correlation between self-assessment scores and change in circulating IGF-I Preparation for Competition 0 –4 –50 –100 –150 –200 –250 r=0.85 –3 –2 –1 80 when IGF-I levels normalized, emphasizing their potential contributing role for the training-­ associated change in IGF-I. Tapering training intensity prior to the competition is a well-known training methodology used to help the athlete to achieve their best performance (i.e., increased rest leading to a psychophysiological restoration) [57]. Interestingly, this strategy is indeed associated with a parallel increase in circulating IGF-I levels. Therefore, measurements of IGF-I may assist coaches and athletes in their training preparations and provide a clue whether the tapering is being effective. Interestingly, in sports that do not plan their training for a specific targeted date of peak performance, like many of the team sports that train in the same relative intensity throughout a regular season (e.g., handball, soccer, etc.), changes in IGF-I level and its major binding protein IGFBP-3 are not typically found [58]. In optimal conditions, during the tapering of training intensity, IGF-I level will increase above baseline levels and will be associated with improved performance; however, this does not occur always. Since IGF-I can be reduced by nutritional imbalance and weight loss, it is possible that a deliberate decrease in body weight in athletes who participate in weight category sports (e.g., judo, wrestling), or even in team sport players prior to major tournaments, may prevent further increase in this anabolic hormone and will be associated “only” with a significant return to baseline values [56, 59]. This emphasizes the importance of proper nutritional counseling all throughout the training season. Previous studies demonstrated in athletes a training-associated negative correlation between circulating IGF-I and ghrelin, a hormone that is secreted by the stomach and pancreas and known to stimulate hunger [60]. Moreover, decreases of ghrelin and leptin, both known to mediate energy balance, were found following a 3-month preseason ­preparatory training in young female handball and basketball players [61]. All together this suggests that hormonal relationships, as one would expect, play a mediating role in training-induced associated energy balance, appetite, body composition, and muscle performance changes. A. Eliakim and D. Nemet Interestingly, despite decreases in circulating IGF-I during period of intense training, physical fitness may still improve, as muscle mass does [62–65]. This suggests that while changes in circulating IGF-I are good markers of the general condition and energy balance of the athlete, they are not necessarily good indicators of the athlete’s performance level. Probably, it is the local muscle levels of these hormones, and their autocrine or paracrine secretion, that are or could be more indicative of skeletal muscle performance [66, 67]. Nonetheless, tapering of the training intensity, was found to be associated with both increased IGF-I level and with further improvement of exercise performance of the athletes [57, 68]. It is still unknown what should be the below baseline permitted decrease of IGF-I levels during periods of heavy training, or what should be the optimal increase of this substance during periods of tapering and reduced training intensity (i.e., what magnitude of change is detrimental versus beneficial). However, the inability to increase circulating IGF-I levels before the target competition may suggest inappropriate recovery and suggest to the athlete and his/her coach that the athlete’s general condition is not optimal. Collection of baseline and training-related hormonal changes, with a comparison to the hormonal response in previous seasons, and the knowledge and experience of the past success may prove to be of a very significant relevance as well. I GF-I Genetics, Sports Selection, and Sports Excellence The potential use of genetic single nucleotide polymorphisms (SNPs) of hormone genes, as a tool to assist in predicting future athletic performance, is currently an extremely challenging topic, mainly because each possible gene makes only a small contribution to the overall heritability. The majority of previous reports of hormonal gene polymorphism and athletic performance in professional athletes studied variations in the IGF-I polymorphism. The polymorphism of IGF-I promoter frequency Exercise and the GH-IGF-I Axis AA AG GG 100 80 60 40 20 ift W ei gh tl um t/J in Sp r ur an ce p 0 En d was significantly greater in athletes (9.2%) compared to controls (2.4%) and in particular among strength (11%) compared to athletes participating in team sports (7.8%) [69]. Our research group previously demonstrated [70] a higher frequency of the IGF-­IC1245T T/T IGF-I promoter polymorphism among Israeli athletes (4.8%), compared to controls (nonexistence). Interestingly, while T/T polymorphism carriers were both endurance and power athletes, endurance athletes were of a national level, but the power athletes were top-­level international and Olympic athletes. This suggests that the IGF1 T/T polymorphism may be more beneficial for power sports performance at the elite level. Along these lines, we also recently assessed [71] the frequency of another polymorphism of the IGF-I gene (i.e., IGF-­Irs7136446) and demonstrated that the frequency of carrying the GG genotype was significantly greater among sprinters compared to weight lifters (see findings depicted in Fig. 5.5). Taken all together, this may suggest that among certain power sports activities, the IGF-I polymorphism is more important for speed rather than strength. In addition, we showed that the IGF2 (rs680) GG genotype frequency was significantly greater among sprinters compared to weight lifters [72], suggesting that carrying this IGF2 polymorphism may also be beneficial mainly for speed-related and not for strength sports. Circulating IGF2 levels were lower among individuals homozygous for the G allele [73], and higher levels of plasma IGF-I were found in individuals carrying the IGF2 GG genotype [74]. It is possible that the beneficial effect of the IGF2 rs680 polymorphism on speed performance is not necessarily mediated through its influence on circulating IGF2, but via its effect on IGF-I levels. This point, however, needs much further investigation. Interestingly, it has been previously demonstrated that in contrast to elite track and field athletes, single nucleotide polymorphisms of IGF1, IGF1 receptor, and IGF2 were not frequent among swimmers [72, 75, 76]. These results possibly suggest that the insulin-like growth factor system is less significant for elite swimming than 81 % prevalence 5 Fig. 5.5 The prevalence of the A/G IGF-I rs7136446 polymorphism among national and top-level Israeli endurance athletes, short-distance runners/jumpers and among weight lifters (p = 0.036, for GG genotype frequency, sprinters vs weight lifters) for running performance. The mechanism for this discrepancy is currently unknown and in need of study. A possible explanation is that swimming excellence is mainly affected by the swimmer’s physical attributes (particularly limb length) and swimming technique [77], possibly masking physiologic, metabolic, and muscle mass differences and enabling tall and technically skilled swimmers to excel in the majority of swimming distances. These results indicate that extreme caution should be done before pooling different types of sport in genetic research because despite seemingly similar metabolic characteristics, ­ athletes from different sport disciplines carry ­ different genetic polymorphisms. Whether a multipotent athlete who wants to develop a competitive career and carries a beneficial IGF system polymorphism should prefer track and field over swimming is currently speculative and hence must be interpreted with caution. Moreover, one should always keep in mind that while a favorable genetic predisposition is essential, psychological features and environmental aspects, including training equipment and facilities, nutrition, familial support, and motivational issues, are also critically essential for top-level sports performance success. 82 Summary In recent years there has been a significant research progress in the field of exercise endocrinology. It is now clear that monitoring changes in the balance of anabolic (GH→IGF-I system) and catabolic hormones and related inflammatory mediators following different types of exercise training and during different stages of the training season may help elite athletes and their coaching staff in developing a more “optimal” training program and in the preparation for competition. In addition, the use of hormonal genetic polymorphisms may serve as an additional assisting tool for talent identification and sports selection and perhaps also for building effective (i.e., more precise) training programs for athletes. Further research is needed, however, to better clarify the complex relationship of training, hormonal responses, nutrition, genetics, and optimal athletic performance in competitive sports. References 1. Krebs JM, Schneider VS, Evans H, Kuo MC, LeBlanc AD. Energy absorption, lean body mass, and total body fat changes during 5 weeks of continuous bed rest. Aviat Space Environ Med. 1990;61(4):314–8. 2. Theintz GE, Howald H, Weiss U, Sizonenko PC. Evidence for a reduction of growth potential in adolescent female gymnasts. J Pediatr. 1993;122(2):306–13. 3. Urhausen A, Kindermann W. The endocrine system in overtraining. In: Warren MP, Constantini NW, editors. Sports endocrinology. New Jersey: Humana Press; 2000. p. 347–70. 4. LeRoith D, Roberts CT Jr. Insulin-like growth factors and their receptors in normal physiology and pathological states. J Pediatr Endocrinol. 1993;6(3–4):251–5. 5. Eliakim A, Nemet D, Cooper DM. Exercise, training and the GH-->IGF-I axis. In: Kraemer WJ, Rogol AD, editors. The endocrine system in sports and exercise. The Encyclopaedia of sports medicine. 11. 1st ed. Oxford, UK: Wiley-Blackwell; 2005. p. 165–79. 6. Nemet D, Rose-Gottron CM, Mills PJ, Cooper DM. Effect of water polo practice on cytokines, growth mediators, and leukocytes in girls. Med Sci Sports Exerc. 2003;35(2):356–63. 7. Nemet D, Oh Y, Kim HS, Hill M, Cooper DM. Effect of intense exercise on inflammatory cytokines and growth mediators in adolescent boys. Pediatrics. 2002;110(4):681–9. A. Eliakim and D. Nemet 8. Leung DW, Spencer SA, Cachianes G, Hammonds RG, Collins C, Henzel WJ, et al. Growth hormone receptor and serum binding protein: purification, cloning and expression. Nature. 1987;330(6148):537–43. 9. Berelowitz M, Szabo M, Frohman LA, Firestone S, Chu L, Hintz RL. Somatomedin-C mediates growth hormone negative feedback by effects on both the hypothalamus and the pituitary. Science. 1981;212(4500):1279–81. 10. Rajaram S, Baylink DJ, Mohan S. Insulin-like growth factor-binding proteins in serum and other biological fluids: regulation and functions. Endocr Rev. 1997;18(6):801–31. 11. Clark RG, Jansson JO, Isaksson O, Robinson IC. Intravenous growth hormone: growth responses to patterned infusions in hypophysectomized rats. J Endocrinol. 1985;104(1):53–61. 12. Mauras N, Blizzard RM, Link K, Johnson ML, Rogol AD, Veldhuis JD. Augmentation of growth hormone secretion during puberty: evidence for a pulse amplitude-modulated phenomenon. J Clin Endocrinol Metab. 1987;64(3):596–601. 13. Corpas E, Harman SM, Blackman MR. Human growth hormone and human aging. Endocr Rev. 1993;14(1):20–39. 14. Merimee TJ, Zapf J, Froesch ER. Insulin-like growth factors in the fed and fasted states. J Clin Endocrinol Metab. 1982;55(5):999–1002. 15. Makimura H, Stanley TL, Sun N, Hrovat MI, Systrom DM, Grinspoon SK. The association of growth hormone parameters with skeletal muscle phosphocreatine recovery in adult men. J Clin Endocrinol Metab. 2011;96(3):817–23. 16. Vendelbo MH, Jorgensen JO, Pedersen SB, Gormsen LC, Lund S, Schmitz O, et al. Exercise and fasting activate growth hormone-dependent myocellular signal transducer and activator of transcription-5b phosphorylation and insulin-like growth factor-I messenger ribonucleic acid expression in humans. J Clin Endocrinol Metab. 2010;95(9):E64–E8. 17. Buckler JM. Exercise as a screening test for growth hormone release. Acta Endocrinol. 1972;69(2):219–29. 18. Sauro LM, Kanaley JA. The effect of exercise duration and mode on the growth hormone responses in young women on oral contraceptives. Eur J Appl Physiol. 2003;90(1–2):69–75. 19. Schwarz AJ, Brasel JA, Hintz RL, Mohan S, Cooper DM. Acute effect of brief low- and high-intensity exercise on circulating insulin-like growth factor (IGF) I, II, and IGF-binding protein-3 and its proteolysis in young healthy men. J Clin Endocrinol Metab. 1996;81(10):3492–7. 20. Eliakim A, Brasel JA, Cooper DM. GH response to exercise: assessment of the pituitary refractory period, and relationship with circulating components of the GH-IGF-I axis in adolescent females. J Pediatr Endocrinol Metab. 1999;12(1):47–55. 21. Cappon J, Brasel JA, Mohan S, Cooper DM. Effect of brief exercise on circulating insulin-like growth factor I. J Appl Physiol. 1994;76(6):2490–6. 5 Exercise and the GH-IGF-I Axis 22. Ronsen O, Haug E, Pedersen BK, Bahr R. Increased neuroendocrine response to a repeated bout of endurance exercise. Med Sci Sports Exerc. 2001;33(4):568–75. 23. Kanaley JA, Weltman JY, Veldhuis JD, Rogol AD, Hartman ML, Weltman A. Human growth hormone response to repeated bouts of aerobic exercise. J Appl Physiol. 1997;83(5):1756–61. 24. Stokes KA, Sykes D, Gilbert KL, Chen JW, Frystyk J. Brief, high intensity exercise alters serum ghrelin and growth hormone concentrations but not IGF-I, IGF-II or IGF-I bioactivity. Growth Hormon IGF Res. 2010;20(4):289–94. 25. Headley SA, Henry K, Nindl BC, Thompson BA, Kraemer WJ, Jones MT. Effects of lifting tempo on one repetition maximum and hormonal responses to a bench press protocol. J Strength Cond Res. 2011;25(2):406–13. 26. Kubukeli ZN, Noakes TD, Dennis SC. Training techniques to improve endurance exercise performances. Sports Med. 2002;32(8):489–509. 27. Meckel Y, Nemet D, Bar-Sela S, Radom-Aizik S, Cooper DM, Sagiv M, et al. Hormonal and inflammatory responses to different types of sprint interval training. J Strength Cond Res. 2011;25(8):2161–9. 28. Meckel Y, Eliakim A, Seraev M, Zaldivar F, Cooper DM, Sagiv M, et al. The effect of a brief sprint interval exercise on growth factors and inflammatory mediators. J Strength Cond Res. 2009;23(1):225–30. 29. Pedersen BK, Steensberg A, Fischer C, Keller C, Ostrowski K, Schjerling P. Exercise and cytokines with particular focus on muscle-derived IL-6. Exerc Immunol Rev. 2001;7:18–31. 30. Eliakim A, Oh Y, Cooper DM. Effect of single wrist exercise on fibroblast growth factor-2, insulin-like growth factor, and growth hormone. Am J Physiol Regul Integr Comp Physiol. 2000;279(2):R548–R53. 31. Nemet D, Hong S, Mills PJ, Ziegler MG, Hill M, Cooper DM. Systemic vs. local cytokine and leukocyte responses to unilateral wrist flexion exercise. J Appl Physiol. 2002;93(2):546–54. 32. Falk B, Eliakim A. Endocrine response to resistance training in children. Pediatr Exerc Sci. 2014;26(4):404–22. 33. Nindl BC, Hymer WC, Deaver DR, Kraemer WJ. Growth hormone pulsatility profile characteristics following acute heavy resistance exercise. J Appl Physiol (1985). 2001;91(1):163–72. 34. Eliakim A, Portal S, Zadik Z, Rabinowitz J, dler-­ Portal D, Cooper DM, et al. The effect of a volleyball practice on anabolic hormones and inflammatory markers in elite male and female adolescent players. J Strength Cond Res. 2009;23(5):1553–9. 35. Nemet D, Portal S, Zadik Z, Pilz-Burstein R, Adler-­ Portal D, Meckel Y, et al. Training increases anabolic response and reduces inflammatory response to a single practice in elite male adolescent volleyball players. J Pediatr Endocrinol Metab. 2012;25(9–10):875–80. 36. Eliakim A, Portal S, Zadik Z, Meckel Y, Nemet D. Training reduces catabolic and inflammatory 83 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. response to a single practice in female volleyball players. J Strength Cond Res. 2013;27(11):3110–5. Kumari M, Tabassum F, Clark C, Strachan D, Stansfeld S, Power C. Social differences in insulin-­ like growth factor-1: findings from a British birth cohort. Ann Epidemiol. 2008;18(8):664–70. Bogin B, Hermanussen M, Blum WF, Assmann C. Sex, sport, IGF-1 and the community effect in height hypothesis. Int J Environ Res Public Health. 2015;12(5):4816–32. Eliakim A, Cooper DM, Nemet D. The GH-IGF-I response to typical field sports practices in adolescent athletes: a summary. Pediatr Exerc Sci. 2014;26(4):428–33. Barnett A. Using recovery modalities between training sessions in elite athletes: does it help? Sports Med. 2006;36(9):781–96. Wilcock IM, Cronin JB, Hing WA. Physiological response to water immersion: a method for sport recovery? Sports Med. 2006;36(9):747–65. Nemet D, Meckel Y, Bar-Sela S, Zaldivar F, Cooper DM, Eliakim A. Effect of local cold-pack application on systemic anabolic and inflammatory response to sprint-interval training: a prospective comparative trial. Eur J Appl Physiol. 2009;107(4):411–7. Collier SR, Casey DP, Kanaley JA. Growth hormone responses to varying doses of oral arginine. Growth Hormon IGF Res. 2005;15(2):136–9. Collier SR, Collins E, Kanaley JA. Oral arginine attenuates the growth hormone response to resistance exercise. J Appl Physiol. 2006;101(3):848–52. Galassetti P, Larson J, Iwanaga K, Salsberg SL, Eliakim A, Pontello A. Effect of a high-fat meal on the growth hormone response to exercise in children. J Pediatr Endocrinol Metab. 2006;19(6):777–86. Cappon JP, Ipp E, Brasel JA, Cooper DM. Acute effects of high fat and high glucose meals on the growth hormone response to exercise. J Clin Endocrinol Metab. 1993;76(6):1418–22. Foster EB, Fisher G, Sartin JL, Elsasser TH, Wu G, Cowan W, et al. Acute regulation of IGF-I by alterations in post-exercise macronutrients. Amino Acids. 2011;42:1405. Willoughby DS, Stout JR, Wilborn CD. Effects of resistance training and protein plus amino acid supplementation on muscle anabolism, mass, and strength. Amino Acids. 2007;32(4):467–77. Ballard TL, Clapper JA, Specker BL, Binkley TL, Vukovich MD. Effect of protein supplementation during a 6-mo strength and conditioning program on insulin-­ like growth factor I and markers of bone turnover in young adults. Am J Clin Nutr. 2005;81(6):1442–8. Eliakim A, Beyth Y. Exercise training, menstrual irregularities and bone development in children and adolescents. J Pediatr Adolesc Gynecol. 2003;16(4):201–6. Ignacio DL, da SSDH, Cavalcanti-de-Albuquerque JP, Louzada RA, Carvalho DP, Werneck-deCastro JP. Thyroid hormone and estrogen regulate A. Eliakim and D. Nemet 84 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. e­ xercise-­induced growth hormone release. PLoS One. 2015;10(4):e0122556. Warren MP, Chua AT. Exercise-induced amenorrhea and bone health in the adolescent athlete. Ann N Y Acad Sci. 2008;1135:244–52. Vanheest JL, Rodgers CD, Mahoney CE, De Souza MJ. Ovarian suppression impairs sport performance in junior elite female swimmers. Med Sci Sports Exerc. 2014;46(1):156–66. Pons V, Riera J, Capo X, Martorell M, Sureda A, Tur JA, et al. Calorie restriction regime enhances physical performance of trained athletes. J Int Soc Sports Nutr. 2018;15:12. Eliakim A, Nemet D, Bar-Sela S, Higer Y, Falk B. Changes in circulating IGF-I and their correlation with self-assessment and fitness among elite athletes. Int J Sports Med. 2002;23(8):600–3. Nemet D, Pontello AM, Rose-Gottron C, Cooper DM. Cytokines and growth factors during and after a wrestling season in adolescent boys. Med Sci Sports Exerc. 2004;36(5):794–800. Steinacker JM, Lormes W, Kellmann M, Liu Y, Reissnecker S, Opitz-Gress A, et al. Training of junior rowers before world championships. Effects on performance, mood state and selected hormonal and metabolic responses. J Sports Med Phys Fitness. 2000;40(4):327–35. Mejri S, Bchir F, Ben Rayana MC, Ben HJ, Ben SC. Effect of training on GH and IGF-1 responses to a submaximal exercise in football players. Eur J Appl Physiol. 2005;95(5–6):496–503. Roemmich JN, Sinning WE. Weight loss and wrestling training: effects on growth-related hormones. J Appl Physiol. 1997;82(6):1760–4. Jurimae J, Cicchella A, Jurimae T, Latt E, Haljaste K, Purge P, et al. Regular physical activity influences plasma ghrelin concentration in adolescent girls. Med Sci Sports Exerc. 2007;39(10):1736–41. Plinta R, Olszanecka-Glinianowicz M, DrosdzolCop A, Chudek J, Skrzypulec-Plinta V. The effect of three-­ month pre-season preparatory period and short-term exercise on plasma leptin, adiponectin, visfatin, and ghrelin levels in young female handball and basketball players. J Endocrinol Investig. 2012;35(6):595–601. Eliakim A, Brasel JA, Mohan S, Barstow TJ, Berman N, Cooper DM. Physical fitness, endurance training, and the growth hormone-insulin-like growth factor I system in adolescent females. J Clin Endocrinol Metab. 1996;81(11):3986–92. Eliakim A, Brasel JA, Mohan S, Wong WL, Cooper DM. Increased physical activity and the growth hormone-­IGF-I axis in adolescent males. Am J Phys. 1998;275(1 Pt 2):R308–R14. Eliakim A, Scheett TP, Newcomb R, Mohan S, Cooper DM. Fitness, training, and the growth hormone- 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. ->insulin-like growth factor I axis in prepubertal girls. J Clin Endocrinol Metab. 2001;86(6):2797–802. Scheett TP, Nemet D, Stoppani J, Maresh CM, Newcomb R, Cooper DM. The effect of endurance-­ type exercise training on growth mediators and inflammatory cytokines in pre-pubertal and early pubertal males. Pediatr Res. 2002;52(4):491–7. Greig CA, Hameed M, Young A, Goldspink G, Noble B. Skeletal muscle IGF-I isoform expression in healthy women after isometric exercise. Growth Hormon IGF Res. 2006;16(5–6):373–6. Zanconato S, Moromisato DY, Moromisato MY, Woods J, Brasel JA, LeRoith D, et al. Effect of training and growth hormone suppression on insulin-like growth factor I mRNA in young rats. J Appl Physiol. 1994;76(5):2204–9. Izquierdo M, Ibanez J, Gonzalez-Badillo JJ, Ratamess NA, Kraemer WJ, Hakkinen K, et al. Detraining and tapering effects on hormonal responses and strength performance. J Strength Cond Res. 2007;21(3):768–75. Krych-Garsztka K, Mizgajska-Wiktor H, Goździcka-­ Józefiak A. An analysis of the regulatory region of the IGF1 gene in professional athletes in youth sports teams. Hum Mov. 2011;12(3):216. Ben-Zaken S, Meckel Y, Nemet D, Eliakim A. Can IGF-I polymorphism affect power and endurance athletic performance? Growth Hormon IGF Res. 2013;23(5):175–8. Ben-Zaken S, Malach S, MeckeL Y, Nemet D, Eliakim A. Frequency of the IGF A/G rs7136446 polymorphism and athletic performance. Acta Kinesiologiae Universitatis Tartuensis. 2016;22:36–46. Ben-Zaken S, Meckel Y, Nemet D, Eliakim A. High prevalence of the IGF2 rs680 GG polymorphism among top-level sprinters and jumpers. Growth Hormon IGF Res. 2017;37:26–30. O'Dell SD, Miller GJ, Cooper JA, Hindmarsh PC, Pringle PJ, Ford H, et al. Apal polymorphism in insulin-­like growth factor II (IGF2) gene and weight in middle-aged males. Int J Obes Relat Metab Disord. 1997;21(9):822–5. Gatford KL, Heinemann GK, Thompson SD, Zhang JV, Buckberry S, Owens JA, et al. Circulating IGF1 and IGF2 and SNP genotypes in men and pregnant and non-pregnant women. Endocr Connect. 2014;3(3):138. Ben Zaken S, Meckel Y, Dror N, Nemet D, Eliakim A. IGF-I and IGF-I receptor polymorphisms among elite swimmers. Pediatr Exerc Sci. 2014;26(4):470–6. Ben-Zaken S, Meckel Y, Nemet D, Eliakim A. IGF-I receptor 275124A>C (rs1464430) polymorphism and athletic performance. J Sci Med Sport. 2015;18(3):323–7. Maglischo EW. Swimming fastest. Champaign: Human Kinetics; 2003. 6 Exercise and Thyroid Function Dorina Ylli, Joanna Klubo-Gwiezdzinska, and Leonard Wartofsky Introduction Thyroid hormone receptors are present in virtually every tissue in the body, thereby permitting an important physiologic role for the two thyroid hormones, thyroxine (T4) and triiodothyronine (T3). Skeletal and cardiac muscle function, pulmonary performance, metabolism, and the neurophysiologic axis are only a few of the important areas affected by thyroid hormone level [1]. Any abnormality in thyroid function causing either an excess or deficiency in circulating thyroid hormone levels can lead to changes in body function at rest and during exercise. The presence of thyroid disease can have a major impact on exercise tolerance resulting in reduced performance of strenuous activities. On the other hand, exercise itself may have direct or indirect effects on thyroid function, either secondary to acute alterations in the integ- D. Ylli MedStar Health Research Institute, Thyroid Cancer Research Center, Washington, DC, USA J. Klubo-Gwiezdzinska National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Disease/ Metabolic Disease Branch, Bethesda, MD, USA L. Wartofsky (*) Thyroid Cancer Research, Georgetown University School of Medicine, MedStar Health Research Institute, Department of Endocrinology, Washington, DC, USA e-mail: leonard.wartofsky@medstar.net rity of the pituitary thyroid axis or to more longlasting changes. In well-trained athletes, alterations in thyroid function can be viewed as an adaptive mechanism associated with enhanced performance possibly serving to provide a better balance between energy consumption and expenditure. Underlying energy balance does appear to play an important role in the effects that exercise may have on the hypothalamus–pituitary–thyroid axis. Reports in the literature indicate that athletes with excessive weight loss may exhibit a “low T3 syndrome” accompanied by amenorrhea (in women) as well as other alterations in pituitary function [2]. Fortunately, thyroid diseases usually can be treated effectively, and most individuals with thyroid disorders should expect to obtain resolution of their thyroid-related symptoms, including those associated with a negative impact on their exercise tolerance. The track athlete, Gail Devers, who has been very public about her experience with Graves’ disease, is a well-known sprinter who went on to win Olympic fame following treatment for her Graves’ disease and may act as a case in point. After a brief overview of normal thyroid physiology, this chapter will provide a survey of the literature describing effects of abnormal thyroid hormone levels on exercise tolerance, with a special focus on alterations in cardiac, muscle, and respiratory function. The chapter will conclude with a review of existing data on the response of the pituitary–thyroid axis to varying levels and types of exercise. © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_6 85 D. Ylli et al. 86 Thyroid Physiology All steps in thyroid hormone (TH) biosynthesis are driven by thyrotropin (TSH) and are intimately linked to iodine metabolism. Dietary iodine is reduced to iodide, is absorbed by the small intestine, and then enters the circulation. Iodide “trapped” by the thyroid gland subsequently undergoes oxidation by thyroid peroxidase (TPO), iodinating tyrosyl residues in the storage protein, thyroglobulin, to form the iodothyronines, monoiodotyrosine (MIT), and diiodotyrosine (DIT). MIT and DIT molecules can then couple to form either tetraiodothyronine (T4) or triiodothyronine (T3), which are the two major thyroid hormones. T4 and T3 are bound within thyroglobulin and stored in thyroid follicles. Under control of TSH, thyroglobulin undergoes endocytosis and proteolytic digestion, releasing T4 and T3 into the circulation. The feedback loop is completed at the hypothalamic level where declining levels of circulating T4 or T3 will prompt secretion of thyrotropin-releasing hormone (TRH), which stimulates synthesis and secretion of TSH. After binding to its specific receptor on the thyroid cell membrane, TSH leads to stimulation of T4 and T3 production. Only 20% of circulating T3 is derived from thyroid secretion, whereas 80% is derived from the monodeiodination of T4 by 5′-deiodinase (type I and type II) in the periphery (see Fig. 6.1) [3]. Since T3 is some 10–15 times more biologically potent than T4, this latter conversion has been termed the “activating” pathway of thyroid hormone metabolism. Alternatively, in certain physiologic and pathologic states, the deiodination of T4 proceeds via a 5-deiodinase (type I and type III), which leads instead to reverse T3 (rT3). Since rT3 is a biologically inactive compound [3], this route of metabolism has been termed the “inactivating” pathway. A precise metabolic role for rT3 has not been described, but diversion of T4 metabolism from the activating to the inactivating pathway serves a nitrogen-sparing and protective effect for the body during times of stress and has been viewed as homeostatic. After binding to a cellular receptor, the thyroid hormones have both genomic and nongenomic effects, the former leading to modulation in expression of nuclear actions, whereas the latter appears to involve plasma membrane/mitochondrial responses [4] (Table 6.1). hyroid Hormone Effects T Hyper- and hypothyroidism, associated with either excess or deficiency of TH, respectively, may have a negative impact on exercise performance. Although TH has pervasive effects on virtually Triiodothyronine (T3) HO Thyroxine (T4) O CH2— CH— COOH 5' deiodinase HO O NH2 CH2— CH — COOH NH2 5 deiodinase HO O CH2— CH— COOH NH2 Reverse triiodothyronine (rT3) Fig. 6.1 Thyroxine, triiodothyronine, and reverse triiodothyronine 6 Exercise and Thyroid Function Table 6.1 Genomic and nongenomic actions of thyroid hormones Genomic actions of thyroid hormones -Positive regulation Sarcoplasmic reticulum calcium adenosine triphosphatase Myosin heavy chain α β1-adrenergic receptors Sodium/potassium adenosine triphosphatase Voltage-gated potassium channels (Kv1.5, Kv4.2, Kv4.3) Adenine nucleotide translocator 1 -Negative regulation T3 nuclear receptor α1 Myosin heavy chain β Phospholamban Sodium/calcium exchanger Adenylyl cyclase types V,VI Nongenomic actions of thyroid hormones Conductivity of sodium, potassium, and calcium channels Actin polymerization status Activation of PI3K/Akt/mTOR signaling pathway Deiodination and decarboxylation of T4 resulting in thyronamine synthesis all functions of the body, the following discussion emphasizes thyroid-related influences on exercise tolerance as mediated via involvement with cellular metabolism and the function of skeletal muscle and the cardiac, vascular, and pulmonary systems. ardiovascular Effects of Thyroid C Hormones Cardiac performance is dependent on the contractility of the heart as well as systemic vascular resistance. Resting tachycardia is very common in hyperthyroidism, and many patients complain of having a “racing” or “pounding” heart. The heart, being itself a muscle, is affected by thyroid hormone levels as is skeletal muscle. The heart relies mainly on serum T3 because there is no significant myocyte intracellular deiodinase activity [5]. TH can affect cardiac action via direct genomic and nongenomic effects on cardiac myocytes and hemodynamic alterations in the periphery that result in increased cardiac filling and modification of cardiac contraction [6]. TH mediates the expression of both struc- 87 tural and regulatory genes in the cardiac myocyte [5]. Thyroid hormone-responsive cardiac genes include sarcoplasmic reticulum calcium/ adenosinetriphosphatase ([Ca2+]/ATPase) and its inhibitor phospholamban, which are involved in regulation of calcium uptake by the sarcoplasmic reticulum during diastole [7], α- and β-myosin heavy chains, the ion channels coordinating the electrochemical responses of the myocardium: sodium/potassium ATPase (Na+/K+-ATPase), voltage-gated potassium channels (Kv1.5, Kv4.2, Kv4.3), and sodium/calcium exchanger [6]. TH increases the expression of β1-adrenergic receptors and downregulates TRα1 receptors [8, 9]. In summary, the genomic action of TH on the heart involves genes which are largely responsible for enhanced contractile function and diastolic relaxation. Thus, T3 markedly shortens diastolic relaxation, i.e., the hyperthyroid heart relaxes with a higher speed (lusitropic activity), whereas diastole is prolonged in hypothyroid states. The nongenomic effects of TH on the cardiac myocyte and on the systemic vasculature tend to occur rapidly. Schmidt et al. documented that T3-enhanced myocardial contractility and reduced systemic vascular resistance occur within 3 min [10]. These rapid T3-mediated effects include changes in membrane ion channels for sodium, potassium, and calcium; effects on actin polymerization; adenine nucleotide translocator 1 in the mitochondrial membrane; and a variety of intracellular signaling pathways in the heart and vascular smooth muscle cells [11, 12]. The actions on channels may determine set points of myocardial excitability and duration of the action potential and contribute to development of tachyarrhythmias [13]. Additional mechanism of T3 actions observed in vitro includes rapid activation of phosphoinositide 3-kinases (PI3K) leading to protein kinase B (Akt) phosphorylation that in turn translocates to the nucleus and promotes mammalian target of rapamycin (mTOR) phosphorylation [14]. As mTOR is important to regulate ribosomal biogenesis and protein translation, the signaling pathway described in these studies may underlie at least one of the nongenomic mechanisms by which T3 regulates cardiac growth and hypertrophy. D. Ylli et al. 88 Moreover, it has been discovered that deiodination and decarboxylation of T4 could generate a biologically active metabolite, thyronamine, which is characterized by actions opposite to those of TH [15, 16]. It has been demonstrated that thyronamine reduces cardiac output, heart rate, systolic pressure, and coronary flow in isolated heart within minutes [16]. Conceivably, a balance between T3 and thyronamine might be responsible for maintaining cardiac homeostasis. Changes in this equilibrium might contribute to the cardiovascular alterations that occur in patients with thyroid disease [17]. I n Vivo Animal Studies on the Role of Abnormal Thyroid Function in the Regulation of Cardiac Response to Exercise It has been believed that one of the main mechanisms of increased cardiac work during hyperthyroidism was the sensitization to catecholamines. However, Hoit et al. in a study on thyrotoxic baboons refuted a role of βl- or β2-adrenergic receptors in any cardiac response to hyperthyroidism [18]. Interestingly, abnormal cardiac response to exercise has been described as being due to an inefficient use of chemical energy stored in adenosine triphosphate (ATP). In hyperthyroid hearts, a larger fraction of energy goes to heat production, whereas in euthyroid animals more is spent for useful contractile energy. Finally, TH modifies the secretory activity of the heart—i.e., T3 has been found to increase mRNA and protein levels of atrial natriuretic factor [19]. Several studies have indicated overactivation of the renin–angiotensin–aldosterone (RAA) system in hyperthyroid animals, documenting increased plasma renin [20, 21] and upregulated synthesis and secretion of angiotensinogen [22] in hyperthyroid rats. In contrast, the plasma renin activity is reduced in experimental hypothyroidism [20]. There is also an evidence of tissue-­ specific regulation of RAA. TH activates some components of cardiac RAA, and hyperthyroidism can promote an increase in cardiac levels of renin, stimulate Ang II generation [23], and raise levels of AT1 and AT2 receptors [20]. In the heart, Ang II exhibits growth-promoting effects by inducing hypertrophy and fibrosis, mediated by the AT1 receptor [24]. Although most of the effects of Ang II related to cardiac remodeling have been attributed to the AT1 receptor, the AT2 receptor is also involved in the development of some cardiac hypertrophy models [25]. There are several literature reports showing that AT1 receptor blockade and ACE inhibition attenuate or prevent the development of cardiac hypertrophy induced by TH in vivo [21, 26, 27]. Some authors suggest that the mechanism of action of these compounds is associated with the alterations in calcium handling [28], while others suggest that these drugs may inhibit AT1 receptor-induced activation of PI3K/Akt/mTOR pathway [29, 30]. In hyperthyroidism structural remodeling such as hypertrophy, left ventricular fibrosis, myocyte lengthening, chamber dilatation, and decreased relative wall thickness have been observed and have been considered as likely to contribute to global left ventricular functional impairment [31]. Clinical Findings In thyroid disease, cardiac structures and function may remain normal at rest; however, impaired left ventricular (LV) function and cardiovascular adaptation to effort become unmasked during exercise [32]. Hypothyroidism Hypothyroidism has been associated with a decrease in intravascular volume, stroke volume, and cardiac index and an increase in systemic vascular resistance, resulting in diastolic hypertension (Table 6.2) [33]. In patients with transient hypothyroidism owing to thyroidectomy, radionuclide ventriculography and right heart catheterization revealed lower cardiac output, stroke volume, and end-diastolic volume at rest, but increased systemic peripheral resistance [34]. In the same individuals, during exercise, heart rate, cardiac output, end diastolic volume, and stroke volume were higher when the patients were euthyroid than when they were hypothyroid. 6 Exercise and Thyroid Function 89 Table 6.2 Cardiovascular changes observed in hyperand hypothyroidism Heart rate Vascular volume Stroke volume Cardiac output SVR LVEF Rest Exercise Diastolic blood pressure Systolic blood pressure LV pre-ejection period LV ejection time Hyperthyroidism ↑ NC ↑ ↑ ↑ ↓ Hypothyroidism ↓NC ↓ ↓ ↓ ↑NC ↑↓NC ↓ ↓ ↓NC ↓ ↑NC ↑NC ↓NC ↓ ↑ ↓ ↑ ↑ increased, ↓ decreased, NC no change, SVR systemic vascular resistance, LVEF left ventricular ejection fraction, LV left ventricular The baseline LV ejection fraction (LVEF) and peak LVEF were shown to be lower in hypothyroid subjects compared with their euthyroid state, although with exercise, the rise of LVEF in the two states was similar [35]. As assessed by radionuclide-­gated pool ventriculography in a younger group (average age 24 years), there was no noticeable change in LVEF with hypothyroidism, although exercise tolerance did improve after levothyroxine (LT4) replacement [36]. Even hypothyroidism of brief duration of only 10 days was associated with an impaired LVEF response to exercise; LVEF response returned to normal with restoration of the euthyroid state [37]. Of interest, the patients still achieved the same workload in either state. Interesting observations have been found in patients with subclinical hypothyroidism (Sc-HypoT) defined as mild elevations of TSH with normal levels of T4, fT4, T3, and fT3. It has been a matter of investigative interest whether the mild hypofunction associated with subclinical hypothyroidism affected any measureable cardiac parameters. An accurate assessment of left ventricular function performed by Doppler echocardiography in patients with stable Sc-HypoT showed no changes in left ventricle morphology. However, the prolonged isovolumic relaxation time and a reduced early-to-late ratio of the transmitral peak flow velocities are suggestive of impaired diastolic function in the sense of slowed relaxation [38]. In the same study, ten randomly selected patients were re-evaluated after achieving euthyroidism by means of 6 months of LT4 administration. The treatment caused no change in the parameters of left ventricle morphology, whereas it normalized systolic and diastolic function. Interestingly, although systemic vascular resistance was comparable in untreated patients and control subjects, it was significantly decreased after LT4 therapy. Similar findings have been documented by Kahaly et al. [39], who assessed cardiac function on effort and physical exercise capacity showing no abnormalities in various cardiac parameters at rest, either before or after LT4 treatment. However, stroke volume, cardiac index, and peak aortic flow velocity were significantly lower, and the pre-ejection period was significantly prolonged during exercise in the untreated patients versus controls. Other authors confirmed early myocardial dysfunction unveiling a difference in longitudinal systolic and diastolic function reserve indexes during exercise in Sc-HypoT patients compared to controls [40]. However, in a large-scale study, structural changes were not observed when comparing patients with normal TSH with patients with TSH > 5 mIU/L [41]. Tadik et al. performing 3-dimensional echocardiography in 94 subjects observed significantly reduced LV cardiac output and ejection fraction in patients with Sc-HypoT compared to both controls and the same patients 1 year after treatment [42]. Furthermore, when women with Sc-HypoT perform physical activity, a slower HR kinetics (intended as time to reach 63% of the HR at steady state) has been observed in the transition from rest to exercise compared with euthyroid women [43]. Evidence supporting reversible left ventricle diastolic dysfunction in patients with subclinical hypothyroidism was documented employing radionuclide ventriculography [44]. The authors found that the time to peak-filling rate was prolonged in ten patients with Sc-HypoT compared 90 to ten normal control subjects. This accurate index of diastolic function normalized after achieving euthyroidism with LT4 therapy. Abnormal diastolic function may impair coronary flow reserve. Hypothyroid individuals may have a form of reversible coronary dysfunction as found in a study of six patients undergoing stress testing before and after LT4 replacement therapy. Prior to replacement therapy, SPECT scanning revealed notable regional perfusion defects in four of six patients, which resolved within 8 weeks of LT4 therapy [45]. Similarly, Oflaz et al. [46] found that coronary flow reserve was lower in patients with Sc-HypoT than in euthyroid subjects. On the contrary, Owen et al. [47] using stress echocardiography with i.v. dobutamine found no differences in resting global, regional left ventricular function or regional myocardial velocities during maximal dobutamine stress between patients and controls or in patients treated with replacement therapy compared with baseline values. To summarize, the vast majority of clinical studies show impaired LV systolic and diastolic function during exercise in patients with both overt and subclinical hypothyroidism. Hyperthyroidism The effects of hyperthyroidism on cardiac function both during rest and exercise are numerous (see Table 6.2) [33]. In thyrotoxicosis, the extent of the various cardiac responses to excess TH is somewhat dependent on the duration and severity of the disorder. Resting tachycardia, a slow decline in postexercise heart rate (HR), atrial fibrillation, decreased exercise tolerance, and, rarely, congestive heart failure (CHF) are seen in thyrotoxic patients. Cardiac complications from hyperthyroidism tend to occur in patients with a history of prior cardiac disease. Atrial fibrillation, atrial enlargement, and CHF are more common in patients over 60 years old with toxic multinodular goiter. Instead, cardiac valve involvement, pulmonary arterial hypertension, and specific cardiomyopathy are more common in Graves’ disease [48]. Augmented blood volume and blood flow to the skin, muscles, and kidneys are seen and may be owing to vasodilators released D. Ylli et al. secondary to increased cellular respiration [49]. A rise in cellular oxygen consumption leads to a higher demand for oxygen and the need to get fuel to the peripheral tissues [49]. An increase in the velocity of cardiac muscle contraction is present, as well as a rise in myosin ATPase activity [50]. Evaluation of systolic time intervals in thyrotoxic subjects reveals a shortening of the LV pre-ejection period along with quicker LV ejection time and isovolumetric contraction [33, 51]. Kahaly et al. analyzed alterations of cardiovascular function and work capacity using stress echocardiography as well as spiroergometry in subjects with untreated thyroid dysfunction, then again after restoration of euthyroidism. At rest, LVEF, stroke volume, and cardiac indices were significantly increased in hyperthyroidism, but exhibited a blunted response to exercise, which normalized after restoration of euthyroidism. During exercise, negative correlations were found between free T3 (fT3) and diastolic blood pressure, maximal workload, HR, and LVEF. This impaired cardiac response to exercise was specifically apparent in older subjects [52–54]. Of note, combined oral LT4/LT3 overdosage has been reported to cause ST wave depressions with treadmill stress testing that resolve with the euthyroid state [55]. In general, diagnostic treadmill testing is best delayed until patients are euthyroid. “Subclinical” hyperthyroidism (Sc-HyperT) is a term that has been applied to patients with undetectable levels of serum TSH, but with normal levels of T4, fT4, T3, and fT3. In one study, there was no difference in LVEF at rest and exercise between Sc-HyperT and controls, whereas overt hyperthyroid subjects had a reduction in LVEF with exercise, increased HR, and cardiac output at both rest and exercise [56]. Supporting evidence was provided by a study performed in 1112 subjects with a 5-year follow-up in which left ventricular mass divided by height did not differ between subjects with and without Sc-HyperT [57]. However, studies by Kaminski et al. indicated worse physical capacity in subjects with Sc-HyperT and the possibility of improvement after therapy. Compared with results after treat- 6 Exercise and Thyroid Function ment, the end-diastolic and end-systolic volume indexes, stroke volume index, and cardiac index were significantly larger in patients with Sc-HyperT. Stroke volume index was negatively correlated with TSH and positively with fT4 and fT3 values, and cardiac index was positively correlated with fT4 and fT3 levels in Sc-HyperT [58]. Analysis of the Framingham Heart Study revealed that TSH was related to left ventricular contractility in women with TSH < 0.5 mU/L TSH [41]. Furthermore, thicker left ventricular posterior wall, higher HR, and a lower achieved maximum workload have been reported in women with nontoxic multinodular goiter treated with mildly suppressive levothyroxine therapy compared to women not under treatment [59]. To summarize, LVEF, stroke volume and cardiac index, may be greater at rest in hyperthyroidism, but the lack of an increase in LVEF with exercise seems to be a reproducible finding. ffects on Systemic Vascular E Resistance (SVR) TH causes decreased resistance in peripheral arterioles through a direct effect on vascular smooth muscle and decreased mean arterial pressure, which, when sensed in the kidneys, activates the RAA system and increases renal sodium absorption. T3 also increases erythropoietin synthesis, which leads to an increase in red cell mass. The combination of both leads to an increased blood volume and preload. In hyperthyroidism, these effects increase cardiac output by 50–300%, while a 30–50% reduction is seen in hypothyroidism [5]. In the vascular smooth muscle cell, TH-mediated effects are the result of both genomic and nongenomic actions. Nongenomic actions target membrane ion channels and endothelial nitric oxide (NO) synthase, which serves to decrease SVR [60, 61]. Indeed, it was recently reported that the PI3K/Akt signaling pathway plays a role in T3-induced NO production by vascular smooth muscle cells and by endothelial cells [11, 62]. 91 Furthermore, T3 has been shown to inhibit vascular remodeling via the inhibition of the cAMP response element binding protein, a nuclear transcription factor involved in the remodeling process [63]. It seems also that voluntary exercise training can improve long-lasting endothelial dysfunction resulting from transient thyroid hormone deficiency in early life [64]. Clinical Findings Hypothyroidism Vascular control mechanisms may be abnormal in hypothyroidism with blunted vasodilatation secondary to reduced endothelium-dependent vasodilatation [65, 66]. In overt hypothyroidism, arterial compliance is reduced, which leads to increased arterial stiffness with higher central augmentation pressure and lower pulse wave velocities. These abnormalities were reversible with adequate LT4 treatment [67, 68]. However, in subclinical hypothyroidism, the study results have been equivocal. Several studies have not found any association between Sc-HypoT and blood pressure at rest [69–71]. In one cross-­sectional study [69], Sc-HypoT was not associated with increased resting blood pressure. Similar results were observed in the cross-­ sectional Busselton thyroid study [70] that included 105 subjects with Sc-HypoT and 1859 euthyroid controls from Western Australia. On the other hand, two large population-based studies with 5872 [72] and 30,728 [73] subjects reported a modest association between high-­ normal serum TSH levels and resting blood pressure. This observation has been confirmed in other studies, suggesting that mild thyroid ­hormone deficiency also may affect vascular tone [74–77]. Several studies documented an improvement of SVR after LT4 replacement [38, 78]. Endothelial dysfunction in patients with hypothyroidism, borderline hypothyroidism, and those with high-normal TSH values using flow-­ mediated arterial dilation (FMD) has been demonstrated with TSH levels correlating inversely to endothelium-dependent dilatation [77]. Impaired endothelium-dependent 92 vasodilatation as a result of a reduction in nitric oxide availability has been demonstrated in Sc-HypoT by Taddei et al. [79]. Studies have also shown that FMD is associated with plasma osteoprotegerin levels in hypothyroid patients [80]. Osteoprotegerin is a member of the tumor necrosis factor (TNF) receptor family involved in vasculature regulation and related with increased cardiovascular mortality. In vitro studies suggest that TH and TSH are involved in regulation of osteoprotegerin expression [81]. Hyperthyroidism Endothelium-dependent arterial dilatation is increased in hyperthyroid patients and is reversible after subtotal thyroidectomy [82]. Ojamaa et al. [83] demonstrated vascular relaxation due to the action of excess TH on the vascular smooth muscle cells. Conceivably, an inability to lower SVR during exercise in the hyperthyroid state might lead to impaired exercise tolerance [84]. In this regard, phenylephrine administration was associated with an increase in SVR and a decrease in cardiac output not seen in euthyroid subjects [85]. On the contrary, a case-control study of 42 patients with untreated overt hyperthyroidism documented similar systolic and diastolic blood pressures during maximal exercise as in 22 healthy controls. Moreover, no changes in systolic and diastolic blood pressure responses to exercise were observed in these patients after restoration of euthyroidism during 6-month follow­up [52]. Similar findings hold true for the patients with Sc-HyperT. In a recent population-based prospective cohort study, Völzke et al. [86] found that Sc-HyperT is not associated with changes in blood pressure, pulse pressure, or incident hypertension. Some smaller studies have reported similar results [52, 87]. Effects in Muscles TH plays a critical role in maintaining homeostasis and influencing the rate of metabolism and energy expenditure. Skeletal muscles contribute to about 20–30% of resting metabolic rate [88]. D. Ylli et al. TH control the expression of myocyte-specific genes coding for myosin isoforms [32], the Na+– K+ ATPase pumps, and the Ca–ATPase canals of the sarcoplasmic reticulum. This explains the increase of contractility and relaxation of skeletal muscles observed in hyperthyroidism, as opposed to hypothyroidism. In both cases muscle performance is reduced, with accumulation of lactic acid at exercise. This is because of defective pyruvate oxidation and proton expulsion in hypothyroidism and of acceleration of glycolysis in hyperthyroidism. Muscle glycolysis exceeds mitochondrial oxidation enhancing the shunting of pyruvate to lactate, thus leading to an increased lactic acid concentrations resulting in intracellular acidosis. Furthermore, TH increases fast myosin and fast-twitch fibers in skeletal muscle, which are less economic in oxygen utilization during contraction than slow-twitch muscle fibers explaining impaired exercise tolerance. I n Vivo Animal Studies on the Role of Abnormal Thyroid Function in the Regulation of Muscle Response to Exercise Animal studies of hypothyroidism reveal that glycogen levels in muscle appear to be normal to increased at rest, whereas during exercise, muscle utilization of glycogen rises as may lactate production [89, 90]. In hypothyroidism, studies reveal a reduction in flow to the fast-twitch type II fibers of high-­ oxidative type muscles [91] compromising exercise capacity via reduced oxygen delivery and endurance through decreased delivery of blood-­ borne substrates [92, 93]. Additionally, decreased mobilization of free fatty acids (FFA) from adipose tissue leads to reduced lipid delivery to skeletal muscle [94]. After exercise the rate of glycogenolysis exceeded those in controls, showing diminished oxidative capacity resulting in lowering the ATP content. Thus, inadequate fuel utilization may be considered as a factor limiting ability for heavy exercise in hypothyroidism [89] probably triggering compensatory mechanisms in gene expression resulting in a slower striated 6 Exercise and Thyroid Function muscle phenotype [95, 96]. Moreover, in distinction to hypothyroid individuals, muscle blood flow is enhanced in hyperthyroid subjects including fast-twitch sections of muscle [94]. In induced hyperthyroidism, compared to euthyroid control rats, the energy cellular potential was increased during exercise, and it remained higher after the recovery period [97] testifying for an impaired cellular energy. THs also promote expression of peroxisome proliferator-­activated receptor-γ coactivator-1α (PGC-1a), which mediates mitochondrial biogenesis and oxidative capacity in skeletal muscle. Acute exercise increases deiodinase-2 expression in skeletal muscle accelerating conversion of T4 to T3 which induces PGC-1a and its downstream effect on mitochondria [98]. Whether physical activity can be recommended in hyperthyroidism is questionable. The effect of T3-induced thyrotoxicosis on exercise tolerance has been studied, with increases noted in resting oxygen uptake and increased lactic acid levels, protein breakdown, and loss of lean body mass [99]. However, Venditti et al. demonstrated in vivo that moderate training attenuated T3-induced increases in hydrogen peroxide (H2O2) production and, therefore, oxidative damage increasing antioxidant protection and decreasing the reactive oxygen species (ROS) flow from the mitochondria to the cytoplasmic compartment [100]. Another study of leucine supplementation in hyperthyroid rats demonstrated a positive effect in physical performance compared to the non-treated group [101]. Clinical Findings Hypothyroidism Hypothyroidism is characterized by a decrease in Ca2+ uptake and ATP hydrolysis by sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA; see Table 6.3) [102]. At least mild elevations in creatine kinase levels are seen in about 90% of hypothyroid patients [103]. In hypothyroid subjects, the alterations in lipid, protein, and carbohydrate metabolism in muscle may have pronounced effects on muscle function. 93 Table 6.3 Muscle changes observed in hyper- and hypothyroidism Muscle strength Type II fibers Lactate: exercise response Sarcoplasmic reticulum Ca2+ uptake PCr/ Pi ratio—exercise PCr recovery rate Hyperthyroidism ↓ ↑ ↑ Hypothyroidism ↓ ↓ ↑ ↑ ↓ ↑ ↓ ↑ ↓ ↑ increased, ↓ decreased, PCr/Pi phosphocreatine/inorganic phosphate, PCr phosphocreatine Exercise may exacerbate this situation and be associated with rhabdomyolysis [104]. Several cases of rhabdomyolysis have been reported [105, 106], and a relation to a reversible defect in muscle glycogenolysis has been suggested [107]. In Hoffmann’s syndrome, another muscle disorder related to hypothyroidism, abnormalities include increased muscle mass, muscle stiffness and weakness, creatine kinase of as much as >10 times normal levels, and repetitive positive waves on electromyography (EMG) [108]. Resolution of symptoms is expected with thyroid hormone replacement. EMG patterns that can be seen with hypothyroidism include fibrillations, increased polyphasic waves, unusual high-frequency discharges, and reduced motor unit recruitment [108]. An abnormal increase in lactate during exercise but not at rest has been described in subclinical hypothyroidism [109]. It was hypothesized that mitochondrial oxidative dysfunction was present and that this dysfunction worsens with length of disease; glycolysis may exceed pyruvate oxidation explaining the lactate buildup. Phosphorous nuclear magnetic resonance spectroscopy (MRS) has been extensively used to investigate noninvasively the energy metabolism of human muscle. It allows tracking of real-time changes in the relative concentrations of the metabolites that are involved in highenergy phosphate metabolism [110]. A study by Kaminsky et al. performing MRS in hypothyroid women subdivided into either moderate hypothyroidism, subacute thyroid deficiency, or severe/ 94 chronic hypothyroidism demonstrated dysfunction of muscle bioenergetics with even mild TH deficiencies [111]. Khushu et al. documented similar abnormalities in the bioenergetic profile in 32 hypothyroid patients [110]. Similarly, Bose et al. showed shifting of equilibrium of ATP breakdown to ADP and inorganic phosphate (Pi) after exercises confirming impaired oxidative phosphorylation in mitochondria [112]. Haluzik et al. compared metabolic changes in 12 hypothyroid women with those in 6 hyperthyroid and 12 euthyroid women. Compared to healthy subjects, hypothyroidism was associated with significantly decreased noradrenaline and glycerol concentrations, whereas the opposite is applied to hyperthyroid patients. These findings suggest altered adrenergic and lipolytic activities in thyroid disorders [113]. Whether the changes occurring in hypothyroidism are observed in subclinical hypothyroidism has been investigated. Changes in phosphometabolites (increased phosphodiester levels and Pi concentration) were similar in patients with overt hypothyroidism compared to Sc-HypoT. However, impaired muscle oxidative metabolism was not observed in Sc-HypoT patients [114]. Sc-HypoT in 3799 otherwise healthy subjects was associated with a lower resting HR and a significantly lower recovery HR [115]. While Reuters et al. observed no changes in muscle functional capacity in Sc-HypoT, symptoms of cramps, weakness, and myalgia were more frequent compared to controls [116], and a lower HR after exercise was observed [115]. Furthermore, Tanriverdi et al. observed Sc-HypoT subjects to have a higher arterial stiffness and lower physical activity duration with a significant difference in neuromuscular symptoms, muscle strength, and functional exercise capacity assessed by a 6-min walk test [117]. Hyperthyroidism Hyperthyroid subjects also have impairment in cellular respiration and reduced exercise endurance [109]. Excess heat generation from the elevated metabolic activity associated with thyrotoxicosis and secondary hyperthermia may D. Ylli et al. adversely impact heat dissipation during exercise and exercise tolerance. However, despite a baseline temperature increase of 0.5 °C in thyrotoxic subjects, exercise-induced temperature rise has been observed not to differ from that in controls [118]. Reduced duration of action potentials and increased polyphasic potentials can be seen with thyrotoxicosis [119]. Muscle weakness is a common complaint in patients with TH excess, and a variety of investigations have addressed muscle changes secondary to hyperthyroidism. Hyperthyroidism is associated with an increase in fast and a decrease in slow-twitch muscle fibers. Thyrotoxicosis appears to induce an oxidative muscular injury secondary to an increase in mitochondrial metabolism and a decrease in glutathione peroxidase, which may be protective against such injury [120]. Glycogen is lower at baseline in thyrotoxicosis and is utilized at a faster rate with an associated increase of serum lactate [121]. According to studies of Ribeiro et al., glycogen storage in hyperthyroidism can be differently distributed in tissues with lower levels in the heart, liver, and soleus and higher levels in mixed fiber type of gastrocnemius during regular swimming [122]. Thyrotoxic periodic paralysis (TPP) is an unusual complication of hyperthyroidism more typically seen in thyrotoxic Asian subjects, although not exclusively so. Patients with TPP suffer from attacks of para- or quadriplegia incited by exercise, high-carbohydrate meals, or high-salt intake [123]. The muscular function of these patients may appear grossly normal before and between episodes, although some patients have a prodrome of muscle stiffness and aching. The pathophysiology revolves around an imbalance in the Na+/ K+ pump. EMG studies reveal that the muscle has reduced excitability during TPP episodes, and low-amplitude muscle action potentials are seen following a paralytic episode [124]. Decreased compound motor action potential amplitudes are found postexercise in TPP [125] and improve following treatment [126]. Of note, muscle fiber conduction velocity measured in two patients with TPP was within normal limits during paralysis episodes, although muscle strength 6 Exercise and Thyroid Function was reduced by 40% during an attack [127]. A comparison of the electrophysiologic response to prolonged exercise between thyrotoxic patients with and without TPP demonstrated a preexisting latent abnormal excitability of the muscle membrane in TPP [128]. TH regulates muscle membrane excitability by increasing Na+/K+ pump-dependent potassium influx [129]. Adding to our insight into the pathophysiology of TPP is the recent discovery of KCNJ18 gene mutations in a third of TPP patients which alter the function of an inwardly rectifying potassium channel named Kir2.6 [130]. There are also a few case reports documenting rhabdomyolysis as a complication of hyperthyroidism [131–133]. Some authors describe significant metabolic changes in exercising muscle exposed to excess TH. Reduced metabolic efficiency of skeletal muscle energetic with decreased phosphocreatine (PCr) in hyperthyroid patients has been documented by MRS [134]. Under thyrotoxic conditions, ATP is promptly depleted, and myopathy easily develops, as the intramuscular glycogen content decreases due to the suppression of glycogenesis and glycogenolysis. During vigorous exercise, glycogen is rapidly consumed, and ATP consumption by the skeletal muscles increases more than the ATP supply. At that time, the compensatory mechanisms include involvement of purine catabolism as a source of energy [135, 136]. Fukui et al. compared the levels of glycolytic metabolites (lactate and pyruvate) as well as purine metabolites (ammonia and hypoxanthine) in treated and untreated Graves’ disease patients vs. normal controls [137]. The study revealed that glycolysis and purine catabolism were remarkably accelerated in hyperthyroidism and thyrotoxic myopathy could be closely related to the acceleration of purine catabolism, which can be normalized only after long-lasting euthyroidism. Moreover, such acceleration of the purine nucleotide cycle is thought to be in part a protective mechanism against a rapid collapse of the ATP energy balance in exercising muscles of patients with hyperthyroidism [137, 138]. Another important question facing clinicians is the effect of treatment with suppressive doses 95 of LT4 necessary in some patients with differentiated thyroid cancer. Vigario et al. [139] addressed this question and documented that muscle mass was lower in the patients on suppressive LT4 treatment than in euthyroid control subjects, but aerobic training, twice a week, during 3 months partially reversed this deteriorating effect of excess TH on muscle mass. Greater attention should be paid to elderly men with subclinical hyperthyroidism who may have accelerated poor physical performance. Also in euthyroid man, higher FT4 was predictive of a lower Short Physical Performance Battery score at the 3-year follow-up [140]. Effects on Pulmonary Function Performance of any strenuous activity especially of endurance training requires the ability of the respiratory system to augment oxygen utilization. Exercise capacity, the maximal capacity for oxygen consumption (VO2 max), and endurance, the ability to perform prolonged exercise at 75% VO2 max, are the two main components of exercise tolerance [141]. Clinical Findings Large goiters, especially firm, nodular substernal goiters, can cause an extrathoracic tracheal obstruction, which can limit air flow to the lungs [142]. Hypothyroidism Altered TH levels can lead to impairment in optimal pulmonary function. Myxedema or profound hypothyroidism is associated with alveolar hypoventilation related to a reversible reduction in hypoxic ventilatory drive [143]. Reductions in lung volumes are seen and include vital capacity, total lung capacity, functional residual capacity, and expiratory reserve volume, as well as a decrease in diffusing capacity for carbon monoxide (DLCO) [144]. LT4 replacement therapy is associated with resolution of the aforementioned changes, but a concomitant reduction in 96 patient weight may also be an important factor in pulmonary function improvement [145]. Frank respiratory failure is unusual. During exercise, hypothyroid subjects were characterized by reduced forced vital capacity and tidal volume at the anaerobic threshold [146]. Also, the increment of minute ventilation and oxygen uptake was significantly lower. A study in women with subclinical hypothyroidism demonstrated a slower VO2 kinetics (defined as the time needed to reach 63% of change in VO2) in both the onset and recovery of exercise and a higher oxygen deficit compared to euthyroid subjects [147]. Conceivably therefore, it seems that levothyroxine treatment of mild or subclinical hypothyroidism can decrease oxygen uptake, improve minute ventilation and cardiopulmonary exercise performance, and improve the ability in these patients to carry out activities of daily life [148]. Hyperthyroidism Thyrotoxicosis has been implicated as a primary cause of decreased cardiorespiratory exercise tolerance [52, 149, 150]. In hyperthyroidism, already at rest, cardiorespiratory capacity is maximally increased, leading to a limited functional reserve, which may explain the inadequate response of ventilation [53]. Dyspnea on exertion is a common complaint although the causes of this symptom remain unclear and may vary from one patient to another [151]. In hyperthyroidism, the respiratory systems adjust to the increased oxygen demand by increasing respiratory rate and minute ventilation [149]. Alveolar ventilation remains normal, but a rise in dead space ventilation is seen, and also the amount of oxygen diffusion from alveoli to the blood may be reduced during periods of strenuous exercise in thyrotoxicosis [152]. Pulmonary function is dependent on not just intrinsic lung function but also the accessory muscles for respiration. Pulmonary compliance and airway resistance tend to remain unchanged, whereas vital capacity and expiratory reserve volume are reduced, implicating respiratory muscle weakness [153]. Other supporting evidence for respiratory muscle dysfunction in D. Ylli et al. thyrotoxic patients is the reduction of maximal inspiratory and expiratory efforts, which are seen to resolve on restoration of euthyroidism [154]. It appears that ventilation increase beyond the oxygen uptake is related to the dead space ventilation [155]. These changes also appear to resolve with appropriate therapeutic intervention [155]. Furthermore, changes in TH levels modify diaphragmatic function as well as muscle fiber type. Goswami et al. documented a more marked functional weakness of the diaphragm in Graves’ disease during maximal respiratory maneuvers, indicating a diminished diaphragmatic reserve that could cause dyspnea on exertion. These changes were reversible after achieving euthyroidism [156]. With cardiac and muscular function being adversely affected by excess TH, one would postulate that work capacity must be reduced in hyperthyroid individuals. A study of maximum power output in hyperthyroid individuals with measurements of work capacity both while thyrotoxic and then euthyroid revealed a 19% increase from a low maximum power output during the thyrotoxic phase compared to the euthyroid state 3 months later. A subset of patients were retested 12 months later, and maximum power output in comparison to controls was in the low normal range and represented a +13% rise from the 3-month test [157]. Oxygen uptake at maximal effort was low during thyrotoxicosis and did not increase at 3 and 12 months. Net mechanical efficiency was also low at baseline and returned back up to normal only by 12 months. Kahaly et al. showed reduced forced vital capacity, 1-second capacity, and increased respiratory resting oxygen uptake (VO2) rate in hyperthyroid patients compared to euthyroid controls. During exercise, decreased tidal volume at the anaerobic threshold was observed as well as a lowered increment of minute ventilation, VO2, and oxygen pulse [53]. The studies are equivocal in terms of the effect of treatment with suppressive doses of LT4 on exercise capacity. Some studies revealed similar blood pressure, heart rate, VO2, VCO2, and anaerobic threshold response to exercise in LT4-treated patients as in healthy control subjects [158]. Other studies found that ventilation parameters 6 Exercise and Thyroid Function between patients and controls were comparable only at rest, but the patients treated with suppressive doses of LT4 had a worse response to exercise (i.e., lower maximal workload, lower peak VO2, and lower anaerobic threshold) [159]. In conclusion, analysis of respiratory gas exchange showed low efficiency of cardiopulmonary function, respiratory muscle weakness, and impaired work capacity in hyperthyroidism which was reversible with restoration of euthyroidism. xercise and Thyroid Axis Response E Exercise is a stressful situation that challenges body homeostasis, so that the organism has to reestablish a new dynamic equilibrium in order to minimize cell damage. One of the systems affected is the hypothalamic–pituitary–thyroid (HPT) axis [160]. Data demonstrate that voluntary exercise adapts the status of the HPT axis, through pathways that are distinct from those observed during food restriction or repeated stress [161]. Lesmana et al. suggested that alteration in TH signaling (increased TRβ1 expression) and TSH reduction observed in vitro after moderate training can contribute to the metabolic adaptation of skeletal muscle to physical activity [162]. Although the belief that a different normal range for thyroid hormones may apply in athletes compared to healthy nonathletes may be considered, data on the effects of exercise on TH metabolism have been inconsistent or even contradictory (see Table 6.4). These divergent results may be due to differences in the intensity of work, duration of exercise, and frequency and design of the training program and to differences in gender, age, and baseline individual physical status of the subjects. In addition, different duration of studies, timing of sampling after exercise, and methodological factors in hormonal assay and data analysis may also be responsible for the discrepancies. Some studies indicated no major changes in the thyroid axis response to exercise. For example, a study of 26 healthy males, given identical diet and physical activity for a week before the test, revealed an increase in T3, T4, and TSH imme- 97 diately after exercise. However, it seems that the changes were mainly due to hemoconcentration, since they became insignificant after adjustment for hematocrit (Hct) [163]. Another study in subjects undergoing different exercise endurance showed similar results: no significant change in FT4 and a small increase (partially from hemoconcentration) in serum T3 and rT3 [164]. Interestingly, some studies indicate that TSH increases after exercise with the response dissipating with repetitive testing, which was suggested to indicate a psychological influence on the TSH rise [165]. In another study a fT4 increase of 25% was seen postexercise [166], but the increase may have been confounded by assay interference by an associated rise in FFAs. TSH also rose by 41%, but could not be correlated with T4/T3 levels. A rise in TSH was seen with both short-­ duration graded exercise and prolonged exercise, but the latter had a peak of 33% lower than with graded exercise [167]. Another study compared the effect of submaximal and maximal exercise effect on TH levels [168]. Maximal exercise was associated with a decrease in TSH, FT4, and ­stable rT3 and rises in T3 during activity, whereas submaximal exercise was associated with an increase in TSH, but T3, rT3, and FT4 were unchanged. [168]. Also, when comparing intensive exercise intervals with steady-state endurance exercise, Hackney et al. observed that the change in TH levels was present 12 hours post exercise only in the intensive exercise group implying a longer period necessary for TH to return to normal. In both groups an increase in fT3, fT4, and rT3 was present immediately post exercise with a decrease in fT3 and increase of rT3 12 hours post exercise only in the intensive exercise group [169].TH changes in ultradistance and long-distance runners have also been investigated. Hesse et al. studied the effect of three distances of 75 km, 45 km, and marathon (42.2 km) with the subjects performing the 45-km run being slightly older than the other two groups. T4 increased in the 75 km and marathon group and decreased together with T3 in the 45-km group postrace. rT3, measured only in the marathon and 75-km groups, rose in both groups. The authors D. Ylli et al. 98 Table 6.4 Reported changes in hypothalamus–pituitary–thyroid axis in association with exercise Caloric status NA NA NA NA NA NA TSH T4 fT4 T3 fT3 rT3 ↑ ↑ ↑ ↑ NC ↑ ↓ NC ↓ ↑ ↑ ↓ ↑ NC Maximal treadmill exercise Sufficient NC Aerobic exercise Running Running Running Runners Ultradistance Ultradistance Ultradistance Intensive exercise Intensive exercise Steady state exercise Steady state exercise Swimming Swimming Swimming Swimming Ballet High-intensity resistance training High-intensity endurance training Deficient NA NA NA Deficient NA NA NA NA NA NA Chronic endurance exercise Chronic endurance exercise High-altitude exercise NA Reference [165] [166] [173] [173] [188] [181] Exercise type Pre-exercise Ergometry Ergometry Ergometry Ergometry Chronic ergometry [163] [2] [168] [174] [168] [183] [170] [170] [170] [169] [169] [169] [169] [164] [176] [176] [176] [197] [195] [195] [180] [183] [196] ↓ NC ↑ NC ↓ NC NC ↑ NC ↓ ↓ ↑ NC NC NC NC ↑ ↓ NC ↑ ↓ ↓ ↑ ↓ ↑ ↑ ↓ ↑ ↑ ↓ NA NA NA NA NA NA NA ↑ NC ↓ NC ↓ NA ↑ NC NC ↑ NC ↓ NC NC 12 hours post exercise ↑ NC NC NC NC ↑ NC ↓ NC NC ↓ NC ↓ ↓ ↓ ↓ NA NC ↓ NA NC NC Comments Anticipation of exercise Normal TRH stim Untrained athletes Well-trained athletes Glucose infusiona Recreational athletes NC TSH response to TRH Transient changes in TH values reflected transcapillary movements of water Not seen in energy-balanced group ↑ NC Maximal exercise Endurance athletesb versus controls NC Submaximal exercise Prevented by caloric increase 75 kmc ↑ 45 kmc 42.2 kmc ↑ Post exercise ↑ 12 hours post exercise ↑ Post exercise ↑ ↓ ↓ 20oCd 26 °C 32 °C Leptin levels correlated with TSH levels Same group of rowers underwent 3 weeks of resistance and 3 weeks of endurance training Identical twins ↓ Amenorrheice Increased GH/IGF-1 axis and low T3 syndrome T4 thyroxine, T3 triiodothyronine, fT4 free thyroxine, fT3 free triiodothyronine, rT3 reverse triiodothyronine, TSH thyrotropin, TRH TSH-releasing hormone, NA not applicable, NC no change, ↑ increase, ↓ decrease a A glucose infusion blunted the exercise-induced changes of rT3, T3, and T4 b Endurance athletes had balanced increase in T3 production and disposal rates in comparison to active and sedentary men c TSH, T4, and T3 are lower in older runners, whereas faster runners had higher T4 and TSH in relation to slower runners d High TSH with longer cold water exposure e T4/fT4, T3/fT3, and rT3 were lower in exercising amenorrheic versus sedentary group. The eumenorrheic exercise group only has a slightly lower fT4 level, but T4 and T3 were slightly lower than the eumenorrheic sedentary group 6 Exercise and Thyroid Function speculated whether the increase in rT3 might be protective against excess glucose metabolism, especially if intracellular glucose deficiency were present [170]. Semple et al. reported on marathon runners revealed no change in TSH, T4, T3, or rT3 levels before and after the marathon [171]. However, another study revealed an increase in TSH and fT4 post-marathon, with a decrease in fT3 and rise in T4 to rT3 conversion, which was still detectable 22 hours following race completion [172]. The level of training of athletes has been shown to affect the TH response to acute exercise. In one investigation, untrained athletes had a rise in T3, a decrease in rT3, and no change in T4, whereas the well-trained athletes were found to have a rise in rT3, no change in T3, and a decrease in T4 levels. It was hypothesized that the rT3 elevation in well-trained athletes might be adaptive to a more efficient cellular oxidation process [173]. Of note in another study, Rone et al. found an increase in T3 production and turnover in well-trained male athletes in comparison to sedentary men [174]. Following a treadmill stress test, TH levels and TRH simulation revealed responses similar in nature among sedentary subjects, regular joggers, and trained marathon runners [175]. Variation in ambient temperature appears to alter the body’s TH response to exercise. One study looking at TH differences in swimmers exercising in different water temperatures demonstrated that TSH and fT4 rose in the colder water, were unchanged at 26 °C, and fell at the warmer temperature, but T3 levels were not affected [176]. Cold receptors appear to regulate a rise in TRH and TSH level in cold water, and exposure duration may affect the peak TSH with higher levels owing to longer times in the water [177, 178]. The chronic effects on thyroid hormone parameters have also been studied in endurance-­ trained athletes. The results of the studies conflict with regard to whether or not baseline TH levels are shifted in well-trained athletes [179]. Identical twins studied during an observed 93 days endurance training period with stable 99 energy intake had an average 5-kg weight loss (primarily fat) and lower baseline fT3, T3 by the end of the exercise period [180]. A shorter study in recreational athletes over 6 weeks revealed no change in TSH or TSH response to TRH stimulation during exercise although the exercise endurance improved [181]. Also no difference was reported in baseline values for T4, T3, and TSH between endurance athletes and sedentary controls over time [174]. Radioactive iodine uptake (RAIU) may be altered secondary to chronic exercise since a lower thyroid uptake of 123I has been found in regular exercising rats and humans in comparison to sedentary subjects [179]. Energy balance plays a role in the body’s TH response to exercise. Data on the response of TH to fasting or malnutrition [182] suggest that the T3 decrease and rT3 increase could reflect a regulatory mechanism to regulate catabolism and energy expenditure. Of note, T3 and rT3 return to normal with refeeding. Loucks and Heath [183] found a decrease in T3 (–15%) and fT3 (–18%) along with an increase in rT3 (+24%) in healthy women undergoing aerobic exercise testing with low-caloric intake. However, this “low T3 syndrome” was not seen in individuals receiving a normo-caloric diet in balance with their energy expenditure. Other studies demonstrated that the reduction of energy availability from 45 kcal/ kgFFM/day to 10 kcal/kgFFM/day was associated with a decrease in T3 levels in women undergoing 5 days of exercise (see Fig. 6.2a) [184, 185]. Especially in amenorrheic athletes, T3 levels have been found to be lower than in eumenorrheic athletes and sedentary women perhaps suggesting a generalized reduction of the energy-­consuming process (see Fig. 6.2b) [185]. Furthermore, the observed correlation between T3 levels and osteocalcin suggests a possible role in collagen formation and matrix mineralization, thus contributing to the athlete triad characterized as a low energy availability or eating disorder, dysmenorrhea, and low bone density [186]. Interestingly, low-caloric diets high in carbohydrate appear to blunt the drop in T3 compared D. Ylli et al. 100 a b 110 Triiodothyronine (%ES) 110 Triiodothyronine (%45) Fig. 6.2 Triiodothyronine (T3) levels (mean ±SE). (a) Reduction of T3 levels in exercising women after 5 days at energy availabilities of 45, 30, 20, and 10 kcal/kgFFM/day. (b) Amenorrheic athletes (AmA), eumenorrheic athletes (EuA), and sedentary women (EuS). Low T3 levels in the athletes suggest a generalized reduction in the rates of energy consuming processes 100 90 80 70 10 20 30 45 Energy availability (kcal/kgFFM/day) to low-carbohydrate intake [187]. Moreover, glucose infusion has been found to diminish the increase in rT3 and T4 along with decrease in T3 [188]. In a military study, rangers were assessed over 4 days of grueling training in conjunction with sleep and caloric deprivation. The training was associated with an initial increase of TH during the first 24 h. After 4 days of training, there was a gradual decrease in T4, fT4, and T3 (65%), whereas rT3 continued to rise. The group that received a higher caloric intake, and therefore less energy deficiency, had a continued increase in T3 and T4. In the energy-deficient groups, TSH decreased during the first day and remained low throughout the training period. The response of TSH to TRH was reduced in all groups, but much less so in the energy-sufficient group [189]. The detected energy deficiency correlated with a decrease in T3 and increase in rT3 in this study [189]. Hackney et al. have demonstrated that these responses to military exercises and their relation to energy deficiency exist in extreme cold as well as hypoxic environments [190, 191]. Higher-altitude exposure has been shown to be associated with an increase in T4 and fT4 [192]. Furthermore, although Stock et al. reported that exercise at elevated altitudes is also notable for a significant increase in T4 and fT4 with even mild activity [193], not all studies entirely agree with these observations [191]. 100 90 80 70 AmA EuA EuS Subject group Animal studies revealed an increase in serum T3 immediately after exercise, with a gradual decrease thereafter to significantly lower values than in controls. Concomitantly, T4 levels progressively increased, resulting in the T3/T4 ratio being significantly decreased 60 and 120 min after the exercise, indicating impaired T4-to-T3 conversion [194]. Simsch et al. assessed hypothalamic–thyroid axis and leptin concentrations in six highly trained rowers. After 3 weeks of resistance training, a reduction in TSH, fT3, and leptin was found, while fT4 was unchanged. Interestingly, leptin levels correlated with basal TSH levels. In contrast, after 3 weeks of endurance training, a significant increase of TSH was observed. The authors interpreted these data to indicate that depression of the hypothalamic–thyroid axis and leptin is associated with training intensity [195]. Studies of Benso et al. also support the concept of low T3 syndrome as an adaptive mechanism to intense training as was seen in nine male well-­trained climbers studied after climbing Mt. Everest and resulting in a low T3 syndrome with no significant change in ghrelin and leptin despite decrease in body weight [196]. Relative to women, amenorrhea is commonly seen in well-trained female athletes. One study found that amenorrheic subjects had lower T4 and T3 levels than the eumenorrheic groups, but the trained eumenorrheic females had slightly 6 Exercise and Thyroid Function lower T4 and T3 levels than the eumenorrheic nonathletes as well [197]. Of interest, the amenorrheic athletes tended to eat less fat and eat more carbohydrates with a similar caloric intake in comparison to the two other groups with more normal menstrual function. Also, the amenorrheic exercise group trained more hours and more strenuously than the other two groups. Oxygen uptake (VO2) was similar in the trained groups, who also weighed less and had lower body fat. As measured by 31phosphorous magnetic resonance spectroscopy (31P-MRS), inorganic phosphate/ phosphocreatine (Pi/PCr) was not different at rest or at exercise, and pH did not differ at any activity level. However, PCr recovery was substantially faster in the eumenorrheic endurance-trained group than in the eumenorrheic nonathletes and amenorrheic athletes, and the Pi/PCr recovery was only different between the eumenorrheic-­ trained athletes and nonathletes [197]. PCr recovery is related to oxidative metabolism, and the fast recovery in trained eumenorrheic athletes indicates a potentially more efficient metabolism. The other parameters examined for exercise metabolism in these subjects were similar. Contrastingly, in another study, levels of TSH, T3, and T4 were not found to be different in oligomenorrheic heavily trained adolescents versus adolescent athletes without “strenuous” exercise with regular menses [198]. Summary In summary, the thyroid function changes secondary to exercise represent complex physiologic responses, which are difficult to characterize fully. Mitigating factors in the TH response to exercise include age, baseline fitness, nutrition status, ambient temperature, altitude, as well as time, intensity, and type of exercise performed. Another important factor in interpretation of the extant literature is that not all TH blood tests were assessed in every study. Moreover, older studies employed less sensitive assay techniques, whereas various assays have improved over time. The detection of increased FFA in several studies, which may interfere with some TH assays, 101 also cannot be overlooked. However, despite these issues, a review of the literature does reveal certain trends (Table 6.4). One of the more consistent findings is that rT3 tends to increase with exercise especially with associated caloric energy deficiency or ultradistance exercise activities. However, TSH appears to be unaffected by exercise in about 50% of studies with an increase in TSH secondary to cold exposure being a noted exception. T4 was found to increase in 46%, decrease in 26%, and be unchanged in 28% of investigations, although an increase was more typically found with caloric energy deficiency, cold exposure, or ultradistance exercise; fT4 follows a similar pattern to T4. T3 was found to be decreased or be unchanged in 73% of study samples and usually is low with caloric energy deficiency (as in low T3 syndrome); fT3 when measured tended to follow the T3 pattern. Many of the TH changes seen especially in athletes with negative energy balance appeared to be reversed with either a high-carbohydrate intake or even glucose infusion. Although well-­ trained athletes may exhibit an increased production and turnover of T4, baseline TH levels do not appear to be affected substantially by chronic exercise (i.e., endurance). References 1. Wartofsky L. The approach to the patient with thyroid disease. In: Becker KL, editor. Principles and practice of endocrinology and metabolism. 2nd ed. Philadelphia: Lippincott; 1995. p. 278–80. 2. Loucks AB, Callister R. Induction and prevention of low-T3 syndrome in exercising women. Am J Phys. 1993;264:924–30. 3. Leonard JL, Koehrle J. Intracellular pathways of iodothyronine metabolism. In: Braverman LE, Dtiger RD, editors. Werner and Ingbar’s the thyroid. 7th ed. Philadelphia: Lippincott; 1996. p. 125–60. 4. Motomura K, Brent GA. Mechanisms of thyroid hormone action: implications for the clinical manifestation of thyrotoxicosis. Endocrinol Metab Clin N Am. 1998;27:1–19. 5. Klein I, Danzi S. Thyroid disease and the heart. Circulation. 2007;116:1725–35. 6. Kahaly GJ, Dillmann WH. Thyroid hormone action in the heart. Endocr Rev. 2005;26:704–28. 7. Dillmann WH. Cellular action of thyroid hormone on the heart. Thyroid. 2002;12:447–52. D. Ylli et al. 102 8. Bahouth SW, Cui X, Beauchamp MJ, et al. Thyroid hormone induces beta1-adrenergic receptor gene transcription through a direct repeat separated by five nucleotides. J Mol Cell Cardiol. 1997;29:3223–37. 9. Zinman T, Shneyvays V, Tribulova N, et al. Acute, nongenomic effect of thyroid hormones in preventing calcium overload in newborn rat cardiocytes. J Cell Physiol. 2006;207:220–31. 10. Schmidt BM, Martin N, Georgens AC, et al. Nongenomic cardiovascular effects of triiodothyronine in euthyroid male volunteers. J Clin Endocrinol Metab. 2002;87:1681–6. 11. Hiroi Y, Kim H-H, Ying H, et al. Rapid nongenomic actions of thyroid hormone. Proc Natl Acad Sci U S A. 2006;103:14104–9. 12. Davis PJ, Davis FB. Nongenomic actions of thyroid hormone on the heart. Thyroid. 2002;12:459–4665. 13. Wang YG, Dedkova EN, Fiening JP, et al. Acute exposure to thyroid hormone increases Na+ current and intracellular Ca2+ in cat atrial myocytes. J Physiol. 2003;546:491–9. 14. Diniz GP, Carneiro-Ramos MS, et al. Angiotensin type 1 receptor mediates thyroid hormone-induced cardiomyocyte hypertrophy through the Akt/GSK-­ 3beta/mTOR signaling pathway. Basic Res Cardiol. 2009;104:653–67. 15. Scanlan TS, Suchland KL, Hart ME, et al. 3-Iodothyronamine is an endogenous and rapid-­ acting derivative of thyroid hormone. Nat Med. 2004;10:638–42. 16. Chiellini G, Frascarelli S, Ghelardoni S, et al. Cardiac effects of 3-iodothyronamine: a new aminergic system modulating cardiac function. FASEB J. 2007;21:1597–608. 17. Axelband F, Dias J, Ferrão FM, et al. Nongenomic signaling pathways triggered by thyroid hormones and their metabolite 3-iodothyronamine on the cardiovascular system. J Cell Physiol. 2011;226:21–8. 18. Hoit BD, Khoury SF, Shao Y. Effects of thyroid hormone on cardiac beta-adrenergic responsiveness in conscious baboons. Circulation. 1997;96:592–8. 19. Liang F, Webb P, Marimuthu A, Zhang S, Gardner DG. Triiodothyronine increases brain natriuretic peptide (BNP) gene transcription and amplifies endothelin-dependent BNP gene transcription and hypertrophy in neonatal rat ventricular myocytes. J Biol Chem. 2003;278:15073–83. 20. Marchant C, Brown L, Sernia C. Renin–angiotensin system in thyroid dysfunction in rats. J Cardiovasc Pharmacol. 1993;22:449–55. 21. Basset A, Blanc J, Messas E, et al. Renin–angiotensin system contribution to cardiac hypertrophy in experimental hyperthyroidism: an echocardiographic study. J Cardiovasc Pharmacol. 2001;37:163–72. 22. Hong-Brown LQ, Deschepper CF. Effects of thyroid hormones on angiotensinogen gene expression in rat liver, brain, and cultured cells. Endocrinology. 1992;130:1231–7. 23. Kobori H, Ichihara A, Suzuki H, et al. Thyroid hormone stimulates renin synthesis in rats without 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. involving the sympathetic nervous system. Am J Phys. 1997;272:227–32. Bader M, Ganten D. Update on tissue renin–angiotensin systems. J Mol Med (Berl). 2008;86:615–21. D’Amore A, Black MJ, Thomas WG. The angiotensin II type 2 receptor causes constitutive growth of cardiomyocytes and does not antagonize angiotensin II type 1 receptor-mediated hypertrophy. Hypertension. 2005;46:1347–54. Asahi T, Shimabukuro M, Oshiro Y, et al. Cilazapril prevents cardiac hypertrophy and postischemic myocardial dysfunction in hyperthyroid rats. Thyroid. 2001;11:1009–15. Pantos C, Paizis I, Mourouzis I, et al. Blockade of angiotensin II type 1 receptor diminishes cardiac hypertrophy, but does not abolish thyroxininduced preconditioning. Horm Metab Res. 2005;37:500–4. Su L, Dai Y, Deng W, et al. Renin–angiotensin system blocking agents reverse the myocardial hypertrophy in experimental hyperthyroid cardiomyopathy via altering intracellular calcium handling. Zhonghua Xin Xue Guan Bing Za Zhi. 2008;36:744 (Abstract). Kenessey A, Ojamaa K. Thyroid hormone stimulates protein synthesis in the cardiomyocyte by activating the Akt-mTOR and p70S6K pathways. J Biol Chem. 2006;281:20666–772. Kuzman JA, O’Connell TD, Gerdes AM. Rapamycin prevents thyroid hormone-induced cardiac hypertrophy. Endocrinology. 2007;148:3477–84. Weltman NY, Wang D, Redetzke RA, Gerdes AM. Longstanding hyperthyroidism is associated with normal or enhanced intrinsic cardiomyocyte function despite decline in global cardiac ­function. PLoS One. 2012;7(10):e46655. https://doi. org/10.1371/journal.pone.0046655. Kahaly GJ, Kampmann C, Mohr-Kahaly S. Cardiovascular hemodynamics and exercise tolerance in thyroid disease. Thyroid. 2002;12:473–81. Amidi M, Leon DF, DeGroot WJ, et al. Effect of the thyroid state on myocardial contractility and ventricular ejection rate in man. Circulation. 1968;38:229–39. Wieshammer S, Keck FS, Waitzinger J, et al. Left ventricular function at rest and during exercise in acute hypothyroidism. Br Heart J. 1988;60:204–11. Forfar JC, Muir AL, Toft AD. Left ventricular function in hypothyroidism: responses to exercise and beta adrenoceptor blockade. Br Heart J. 1982;48:278–84. Smallridge RC, Goldman MH, Raines K, et al. Rest and exercise left ventricular ejection fraction before and after therapy in young adults with hyperthyroidism and hypothyroidism. Am J Cardiol. 1987;60:929–30. Donaghue K, Hales I, Allwright S, et al. Cardiac function in acute hypothyroidism. Eur J Nucl Med. 1985;11:147–9. Biondi B, Fazio S, Palmieri EA, et al. Left ventricular diastolic dysfunction in patients with sub- 6 Exercise and Thyroid Function 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. clinical hypothyroidism. J Clin Endocrinol Metab. 1999;84:2064–7. Kahaly GJ. Cardiovascular and atherogenic aspects of subclinical hypothyroidism. Thyroid. 2000;10:665–79. Akcakoyun M, Kaya H, Kargin R, Pala S, Emiroglu Y, Esen O, Karapinar H, Kaya Z, Esen AM. Abnormal left ventricular longitudinal functional reserve assessed by exercise pulsed wave tissue Doppler imaging in patients with subclinical hypothyroidism. J Clin Endocrinol Metab. 2009;94(8):2979–83. Pearce EN, Yang Q, Benjamin EJ, Aragam J, Vasan RS. Thyroid function and left ventricular structure and function in the Framingham heart study. Thyroid. 2010;20(4):369–73. Tadic M, Ilic S, Kostic N, Caparevic Z, Celic V. Subclinical hypothyroidism and left ventricular mechanics: a three-dimensional speckle tracking study. J Clin Endocrinol Metab. 2014;99(1): 307–14. Almas SP, Werneck FZ, Coelho EF, Teixeira PF, Vaisman M. Heart rate kinetics during exercise in patients with subclinical hypothyroidism. J Appl Physiol (1985). 2017;122(4):893–8. Brenta G, Mutti LA, Schnitman M, et al. Assessment of left ventricular diastolic function by radio-nuclide ventriculography at rest and exercise in subclinical hypothyroidism, and its response to L-thyroxine therapy. Am J Cardiol. 2003;91:1327–30. Bernstein R, Muller C, Midtbo K, et al. Silent myocardial ischemia in hypothyroidism. Thyroid. 1995;5:443–6. Oflaz H, Kurt R, Cimen A, et al. Coronary flow reserve is also impaired in patients with subclinical hypothyroidism. Int J Cardiol. 2007;120:414–6. Owen PJD, Rajiv C, Vinereanu D, et al. Subclinical hypothyroidism, arterial stiffness and myocardial reserve. J Clin Endocrinol Metab. 2006;9: 2126–32. Biondi B, Kahaly GJ. Cardiovascular involvement in patients with different causes of hyperthyroidism. Nat Rev Endocrinol. 2010;6(8):431–43. Klein I. Thyroid hormone and the cardiovascular system. Am J Med. 1988;88:631–7. Schwartz K, Lecarpenter Y, Martin JL, et al. Myosin isoenzyme distribution correlates with speed of myocardial contraction. J Mol Cell Cardiol. 1981;13:1071–5. Parisi AF, Hamilton BP, Thomas CN, et al. The short cardiac pre-ejection period, an index of thyrotoxicosis. Circulation. 1974;49:900–4. Kahaly GJ, Wagner S, Nieswandt J, et al. Stress echocardiography in hyperthyroidism. J Clin Endocrinol Metab. 1999;84:2308–13. Kahaly GJ, Nieswandt J, Wagner S, et al. Ineffective cardiorespiratory function in hyperthyroidism. J Clin Endocrinol Metab. 1998;83:4075–8. Kahaly GJ, Nieswandt J, Mohr-Kahaly S. Cardiac risks of hyperthyroidism in the elderly. Thyroid. 1998;8:1165–9. 103 55. Peterson CR, Jones RC. Abnormal post-exercise electrocardiogram due to iatrogenic hyperthyroidism. Mil Med. 1969;134:694–7. 56. Foldes J, Istvanffy M, Halmagyi M, et al. Hyperthyroidism and the heart: study of the left ventricular function in preclinical hyperthyroidism. Acta Med Hung. 1986;43:23–9. 57. Dörr M, Ittermann T, Aumann N, Obst A, Reffelmann T, Nauck M, Wallaschofski H, Felix SB, Völzke H. Subclinical hyperthyroidism is not associated with progression of cardiac mass and development of left ventricular hypertrophy in middle-aged and older subjects: results from a 5-year follow-up. Clin Endocrinol. 2010;73(6):821–6. 58. Kaminski G, Dziuk M, Szczepanek-Parulska E, Zybek-Kocik A, Ruchala M. Electrocardiographic and scintigraphic evaluation of patients with subclinical hyperthyroidism during workout. Endocrine. 2016;53:512–9. 59. Di Luigi L, Parisi A, Quaranta F, Romanelli F, Tranchita E, Sgrò P, Nardi P, Fattorini G, Cavaliere R, Pigozzi F, D'Armiento M, Lenzi A. Subclinical hyperthyroidism and sport eligibility: an exploratory study on cardiovascular pre-participation screening in subjects treated with levothyroxine for multinodular goiter. J Endocrinol Investig. 2009;32(10):825–31. 60. Carrillo-Sepúlveda MA, Ceravolo GS, Fortes ZB, et al. Thyroid hormone stimulates NO production via activation of the PI3K/Akt pathway in vascular myocytes. Cardiovasc Res. 2010;85:560–70. 61. Napoli R, Guardasole V, Angelini V, et al. Acute effects of triiodothyronine on endothelial function in human subjects. J Clin Endocrinol Metab. 2007;92:250–4. 62. Kuzman JA, Gerdes AM, Kobayashi S, et al. Thyroid hormone activates Akt and prevents serum starvation-­induced cell death in neonatal rat cardiomyocytes. J Mol Cell Cardiol. 2005;39:841–4. 63. Fukuyama K, Ichiki T, Imayama I, et al. Thyroid hormone inhibits vascular remodelling through suppression of CAMP response element binding protein activity. Arterioscler Thromb Vasc Biol. 2006;26:2049–55. 64. Gaynullina DK, Borzykh AA, Sofronova SI, Selivanova EK, Shvetsova AA, Martyanov AA, Kuzmin IV, Tarasova OS. Voluntary exercise training restores anticontractile effect of NO in coronary arteries of adult rats with antenatal/early postnatal hypothyroidism. Nitric Oxide. 2018;74:10–8. 65. McAllister RM, Delp MD, Laughlin MH. A review of effects of hypothyroidism on vascular transportin skeletal muscle during exercise. Can J Appl Physiol. 1997;22:1–10. 66. Delp MD, McAllister RM, Laughlin MH. Exercise training alters aortic vascular reactivity in hypothyroid rats. Am J Phys. 1995;268:1428–35. 67. Obuobie K, Smith J, Evans LM, et al. Increased central arterial stiffness in hypothyroidism. J Clin Endocrinol Metab. 2002;87:4662–6. 104 68. Dagre AG, Lekakis JP, Papamichael CM, et al. Arterial stiffness is increased in subjects with hypothyroidism. Int J Cardiol. 2005;103:1–6. 69. Duan Y, Peng W, Wang X, et al. Community based study of the association of subclinical thyroid dysfunction with blood pressure. Endocrine. 2009;35:136–42. 70. Walsh JP, Bremner AP, Bulsara MK, et al. Subclinical thyroid dysfunction and blood pressure: a community-­ based study. Clin Endocrinol. 2006;65:486–91. 71. Takashima N, Niwa Y, Mannami T, et al. Characterization of subclinical thyroid dysfunction from cardiovascular and metabolic viewpoints: the Suita study. Circ J. 2007;71:191–5. 72. Iqbal A, Figenschau Y, Jorde R. Blood pressure in relation to serum thyrotropin: the tromso study. J Hum Hypertens. 2006;20:932–6. 73. Asvold BO, Bjoro T, Nilsen TI, et al. Association between blood pressure and serum thyroid-­ stimulating hormone concentration within the reference range: a population- based study. J Clin Endocrinol Metab. 2007;92:841–5. 74. Luboshitzky R, Aviv A, Herer P, et al. Risk factors for cardiovascular disease in women with subclinical hypothyroidism. Thyroid. 2002;12:421–5. 75. Faber J, Petersen L, Wiinberg N, et al. Hemodynamic changes after levothyroxine treatment in subclinical hypothyroidism. Thyroid. 2002;12:319–24. 76. Nagasaki T, Inaba M, Kumeda Y, et al. Increased pulse wave velocity in subclinical hypothyroidism. J Clin Endocrinol Metab. 2006;91:154–8. 77. Lekakis J, Papamichael C, Alevizaki M, et al. Flow-­ mediated, endothelium dependent vasodilatation is impaired in subjects with hypothyroidism, borderline hypothyroidism, and high normal serum thyrotropin (TSH) values. Thyroid. 1997;7:411–4. 78. Yazici M, Gorgulu S, Sertbas Y, et al. Effects of thyroxin therapy on cardiac function in patients with subclinical hypothyroidism: index of myocardial performance in the evaluation of left ventricular function. Int J Cardiol. 2004;95:135–43. 79. Taddei S, Caraccio N, Virdis A, et al. Impaired endothelium-dependent vasodilatation in subclinical hypothyroidism: beneficial effect of levothyroxine therapy. J Clin Endocrinol Metab. 2003;88:3731–7. 80. Xiang G, Sun H, Hou J. Changes in endothelial function and its association with plasma osteoprotegerin in hypothyroidism with exercise induced silent myocardial ischaemia. Clin Endocrinol. 2008;69:799–803. 81. Hofbauer LC, Kluger S, Kuhne CA, et al. Detection and characterization of RANK ligand and osteoprotegerin in the thyroid gland. J Cell Biochem. 2002;86:642–50. 82. Guang-da X, Hong-yan C, Xian-mei Z. Changes in endothelium-dependent arterial dilation before and after subtotal thyroidectomy in subjects with hyperthyroidism. Clin Endocrinol. 2004;61:400–4. D. Ylli et al. 83. Ojamaa K, Klemperer JD, Klein I. Acute effects of thyroid hormone on vascular smooth muscle. Thyroid. 1996;6:505–12. 84. Graettinger JS, Muenster JJ, Selverstone LA, et al. A correlation of clinical and hemodynamic studies in patients with hyperthyroidism with and without congestive heart failure. J Clin Invest. 1959;38:1316–27. 85. Theilen EO, Wilson WR. Hemodynamic effects of peripheral vasoconstriction in normal and thyrotoxic patients. J Appl Physiol. 1967;22:207–10. 86. Völzke H, Ittermann T, Schmidt CO, et al. Subclinical hyperthyroidism and blood pressure in a -population-based prospective cohort study. Eur J Endocrinol. 2009;161:615–21. 87. Kimura H, Kawagoe Y, Kaneko N, et al. Low efficiency of oxygen utilization during exercise in hyperthyroidism. Chest. 1996;110:1264–70. 88. Silva LE. Thermogenic mechanism and their hormonal regulation. Physiol Res. 2006;86:435–64. 89. Kaciuba-Uscilko H, Brzezinska Z, Kruk B, et al. Thyroid hormone deficiency and muscle metabolism during light and heavy exercise in dogs. Pflugers Arch. 1988;412:366–7. 90. Ramsay ID. Muscle dysfunction in hyperthyroidism. Lancet. 1966;2:931–4. 91. McAllister RM, Delp MD, Laughlin MH. Muscle blood flow during exercise in sedentary and trained hypothyroid rats. Am J Phys. 1995;269:949–54. 92. Wieshammer S, Keck FS, Waitzinger J. Acute hypothyroidism slows the rate of left ventricular -diastolic relaxation. Can J Physiol Pharmacol. 1989;67:1007–10. 93. McAllister RM, Ogilvie RW, Terjung RL. Functional and metabolic consequences of skeletal muscle remodeling in hypothyroidism. Am J Phys. 1991;260:272–9. 94. McAllister RM, Sansone JC, Laughlin MH. Effects of hyperthyroidism on muscle blood flow during exercise in the rat. Am J Phys. 1995;268:330–5. 95. Caiozzo VJ, Haddad F. Thyroid hormone: modulation of muscle structure, function, and adaptive responses to mechanical loading. Exerc Sport Sci Rev. 1996;24:321–61. 96. McCarthy JJ, Vyas DR, Tsika GL, et al. Segregated regulatory elements direct beta-myosin heavy chain expression in response to altered muscle activity. J Biol Chem. 1999;274:14270–9. 97. Górecka M, Synak M, Brzezińska Z, Dąbrowski J, Żernicka E. Effect of triiodothyronine (T3) excess on fatty acid metabolism in the soleus muscle from endurance-trained rats. Biochem Cell Biol. 2016;94(2):101–8. 98. Bocco BM, Louzada RA, Silvestre DH, Santos MC, Anne-Palmer E, Rangel IF, Abdalla S, Ferreira AC, Ribeiro MO, Gereben B, Carvalho DP, Bianco AC, Werneck-de-Castro JP. Thyroid hormone activation by type 2 deiodinase mediates exercise-induced peroxisome proliferator-activated receptor-γ coactivator-1α expression in skeletal muscle. J Physiol. 2016;594(18):5255–69. 6 Exercise and Thyroid Function 99. Martin WH, Spina RJ, Korte E, et al. Mechanisms of impaired exercise capacity in short duration experimental hyperthyroidism. J Clin Invest. 1991;88:2047–53. 100. Venditti P, Bari A, Di Stefano L, Di Meo S. Effect of T3 on metabolic response and oxidative stress in skeletal muscle from sedentary and trained rats. Free Radic Biol Med. 2009;46(3):360–6. 101. Fidale TM, Antunes HKM, Roever L, Gonçalves A, Puga GM, Silva RPM, de Resende FN, de Souza FR, Fidale BM, Lizardo FB, Resende ES. Leucine supplementation improves effort tolerance of rats with hyperthyroidism. Front Physiol. 2018;9:1632. 102. Sukp J. Alterations of Ca2+ uptake and Ca2+ -activated ATPase of cardiac sarcoplasmic reticulum in hyper- and hypothyroidism. Biochim Biophys Acta. 1971;252:324–37. 103. Graig FA, Smith JC. Serum creatinine phosphokinase activity in altered thyroid states. J Clin Endocrinol Metab. 1965;25:723–31. 104. Emser W, Schimrigk K. Myxedema myopathy: a case report. Eur Neurol. 1977;16:286. 105. Salehi N, Agoston E, Munir I, Thompson GJ. Rhabdomyolysis in a patient with severe hypothyroidism. Am J Case Rep. 2017;18:912–8. 106. Zhou C, Lai S, Xie Y, Zhang S, Lu Y. Rhabdomyolysis in a patient complicated with hypopituitarism and multiple organ dysfunction syndrome and the literature review. Am J Emerg Med. 2018;36(9):1723. e1–1723.e6. https://doi.org/10.1016/j. ajem.2018.06.019. Epub 2018 Jun 7. 107. Riggs JE. Acute exertional rhabdomyolysis in hypothyroidism: the result of a reversible defect in glycogenolysis. Mil Med. 1990;155:171–2. 108. Klein I, Parker M, Shebert R. Hypothyroidism presenting as muscle stiffness and pseudohypertrophy: Hoffmann’s syndrome. Am J Med. 1981;70:891–4. 109. Monzani F, Caraccio N, Siciliano G, et al. Clinical and biochemical features of muscle dysfunction in subclinical hypothyroidism. J Clin Endocrinol Metab. 1997;82:3315–8. 110. Khushu S, Rana P, Sekhri T, et al. Bio-energetic impairment in human calf muscle in thyroid -disorders: a 31P MRS study. Magn Reson Imaging. 2010;28:683–9. 111. Kaminsky P, Robin-Lherbier B, Brunotte F, et al. Energetic metabolism in hypothyroid skeletal muscle, as studied by phosphorous magnetic resonance spectroscopy. J Clin Endocrinol Metab. 1992;74:124–9. 112. Bose S, French S, Evans FJ, et al. Metabolic network control of oxidative phosphorylation: multiple roles of inorganic phosphate. J Biol Chem. 2003;278:39155–65. 113. Haluzik M, Nedvidkova J, Bartak V, et al. Effects of hypo- and hyperthyroidism on noradrenergic activity and glycerol concentrations in human subcutaneous abdominal adipose tissue assessed with microdialysis. J Clin Endocrinol Metab. 2003;88:5605–8. 105 114. Rana P, Sripathy G, Varshney A, Kumar P, Devi MM, Marwaha RK, Tripathi RP, Khushu S. Phosphorous magnetic resonance spectroscopy-based skeletal muscle bioenergetic studies in subclinical hypothyroidism. J Endocrinol Investig. 2012;35(2):129–34. 115. Maor E, Kivity S, Kopel E, Segev S, Sidi Y, Goldenberg I, Olchovsky D. Differences in heart rate profile during exercise among subjects with subclinical thyroid disease. Thyroid. 2013;23(10):1226–32. 116. Reuters VS, Teixeira Pde F, Vigário PS, Almeida CP, Buescu A, Ferreira MM, de Castro CL, Gold J, Vaisman M. Functional capacity and muscular abnormalities in subclinical hypothyroidism. Am J Med Sci. 2009;338(4):259–63. 117. Tanriverdi A, Ozcan Kahraman B, Ozsoy I, Bayraktar F, Ozgen Saydam B, Acar S, Ozpelit E, Akdeniz B, Savci S. Physical activity in women with subclinical hypothyroidism. J Endocrinol Investig. 2018;42:779. 118. Nazar K, Chwalbinska-Moneta J, Machalla J, et al. Metabolic and body temperature changes during exercise in hyperthyroid patients. Clin Sci Mol Med. 1978;54:323–7. 119. Ramsay ID. Electromyography in thyrotoxicosis. Q J Med. 1965;34:255. 120. Asayama K, Kato K. Oxidative muscular injury and its relevance to hyperthyroidism. Free Radic Biol Med. 1990;8:293–303. 121. Fitts RH, Brimmer CJ, Troup JP, et al. Contractile and fatigue properties of thyrotoxic rat skeletal muscle. Muscle Nerve. 1984;7:470–7. 122. Ribeiro LF, Teixeira IP, Aparecido da Silva G, Dalia RA, Júnior MC, Bertolini NO, Rostom de Mello MA, Luciano E. Effects of swimming training on tissue glycogen content in experimental thyrotoxic rats. Can J Physiol Pharmacol. 2012;90(5):587–93. 123. Gubran C, Narain R, Malik L, Saeed SA. A young man presenting with paralysis after vigorous exercise. BMJ Case Rep. 2012;27:2012. 124. Kelley DE, Garhib H, Kennedy FP, et al. Thyrotoxic periodic paralysis: report of 10 cases and review of the electromyographic findings. Arch Intern Med. 1989;149:2597–600. 125. McManis PG, Lambert EH, Daube JR. The exercise test in periodic paralysis. Muscle Nerve. 1986;9:704–10. 126. Jackson CE, Barohn RJ. Improvement of the exercise test after therapy in thyrotoxic periodic -paralysis. Muscle Nerve. 1992;15:1069–71. 127. Links TP, van der Hoeven JR. Improvement of the exercise test after therapy in thyrotoxic periodic paralysis. Muscle Nerve. 1993;16:1132–3. 128. Arimura K, Arimura Y, Ng AR, et al. Muscle membrane excitability after exercise in thyrotoxic periodic paralysis and thyrotoxicosis without periodic paralysis. Muscle Nerve. 2007;36:784–8. 129. Oh VM, Taylor EA, Yeo SH, et al. Cation transport across lymphocyte plasma membranes in euthyroid and thyrotoxic men with and without D. Ylli et al. 106 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 144. 145. hypokalaemic periodic paralysis. Clin Sci (Lond). 1990;78:199–206. Falhammar H, Thorén M, Calissendorff J. Thyrotoxic periodic paralysis: clinical and molecular aspects. Endocrine. 2013;43(2):274–84. Lichtstein DM, Arteaga RB. Rhabdomyolysis associated with hyperthyroidism. Am J Med Sci. 2006;332:103–5. Alshanti M, Eledrisi MS, Jones E. Rhabdomyolysis associated with hyperthyroidism. Am J Emerg Med. 2001;19:317. Summachiwakij S, Sachmechi I. Rhabdomyolysis induced by nonstrenuous exercise in a patient with graves' disease. Case Rep Endocrinol. 2014;2014:286450. Erkintalo M, Bendahan D, Mattéi JP, et al. Reduced metabolic efficiency of skeletal muscle energetics in hyperthyroid patients evidenced quantitatively by in vivo phosphorus-31 magnetic resonance spectroscopy. Metabolism. 1998;47:769–76. Ruff RL. Endocrine myopathies. In: Engel AG, Banker BQ, editors. Myology. New York: Mc Graw Hill; 1986. p. 1881–7. Zoref-Shani E, Shainberg A, Kessler-Icekson G. Production and degradation of AMP in cultured rat skeletal and heart muscle: a comparative study. Adv Exp Med Biol. 1986;195:485–91. Fukui H, Taniguchi S, Ueta Y, et al. Activity of the purine nucleotide cycle of the exercising muscle in patients with hyperthyroidism. J Clin Endocrinol Metab. 2001;86:2205–10. Hisatome I, Ishiko R, Mashiba H, et al. Excess purine degradation in skeletal muscle with hyperthyroidism. Muscle Nerve. 1990;13:558–9. Vigário PS, De Oliveira CD, Cordeiro MF, et al. Effects of physical activity on body composition and fatigue perception in patients on thyrotropin-­ suppressive therapy for differentiated thyroid carcinoma. Thyroid. 2011;21:695–700. Ceresini G, Ceda GP, Lauretani F, Maggio M, Bandinelli S, Guralnik JM, Cappola AR, Usberti E, Morganti S, Valenti G, Ferrucci L. Mild thyroid hormone excess is associated with a decreased physical function in elderly men. Aging Male. 2011;14(4):213–9. McAllister RM, Delp MD, Laughlin MH. Thyroid status and exercise tolerance: cardiovascular and metabolic considerations. Sports Med. 1995;20:189–98. Bahn RS, Castro MR. Approach to the patient with nontoxic multinodular goiter. J Clin Endocrinol Metab. 2011;96:1202–12. Zwillich CW, Pierson OJ, Hofeldt FD, et al. Ventilatory control in myxedema and hypothyroidism. N Engl J Med. 1975;292:662–5. Ingbar DH. The respiratory system in hypothyroidism. In: Braverman LE, Utiger RD, editors. Werner and Ingbar’s the thyroid. 7th ed. Philadelphia: Lippincott; 1996. p. 805–10. Wilson WR, Bedell ON. The pulmonary abnormalities in myxedema. J Clin Invest. 1960;39:42. 146. Wassermann K. Diagnosing cardiovascular and lung pathophysiology from exercise gas exchange. Chest. 1997;112:1091–101. 147. Werneck FZ, Coelho EF, de Lima JR, Laterza MC, Barral MM, Teixeira Pde F, Vaisman M. Pulmonary oxygen uptake kinetics during exercise in subclinical hypothyroidism. Thyroid. 2014;24(6):931–8. 148. Mainenti MR, Vigário PS, Teixeira PF, Maia MD, Oliveira FP, Vaisman M. Effect of levothyroxine replacement on exercise performance in subclinical hypothyroidism. J Endocrinol Investig. 2009;32(5):470–3. 149. Kahaly G, Hellermann J, Mohr-Kahaly S, et al. Impaired cardiopulmonary exercise capacity in hyperthyroidism. Chest. 1996;109:57–61. 150. Hellermann J, Kahaly GJ. Cardiopulmonary involvement in thyroid disease. Pneumologie. 1996;50:375–80. 151. Small D, Gibbons W, Levy RD, et al. Exertional dyspnea and ventilation in hyperthyroidism. Chest. 1992;101:1268–73. 152. Ayers J, Clark TH, Maisey MN. Thyrotoxicosis and dyspnea. Clin Endocrinol. 1982;164:645. 153. Massey DG, Becklake MR, McKenzie JM, et al. Circulatory and ventilatory response to exercise in thyrotoxicosis. N Engl J Med. 1967;276:1104–12. 154. Siafakas NM, Milona I, Salesiotou V, et al. Respiratory muscle strength in hyperthyroidism before and after treatment. Am Rev Respir Dis. 1992;146:1025–9. 155. Stein M, Kimbel P, Johnson RL. Pulmonary function in hyperthyroidism. J Clin Invest. 1960;40: 348–63. 156. Goswami R, Guleria R, Gupta AK, et al. Prevalence of diaphragmatic muscle weakness and dyspnoea in Graves’ disease and their reversibility with carbimazole therapy. Eur J Endocrinol. 2002;147:299–303. 157. Sestoft L, Saltin B. The low physical working capacity of thyrotoxic patients is not normalized by oral antithyroid treatment. Clin Physiol. 1988;8:9–15. 158. Portella RB, da Costa Silva JL, Wagman MB, et al. Exercise performance in young and middle-aged female patients with subclinical hyperthyroidism. Thyroid. 2006;16:731–5. 159. Mercuro G, Panzuto MG, Bina A, et al. Cardiac function, physical exercise capacity, and quality of life during long-term thyrotropin-suppressive therapy with levothyroxine: effect of individual dose tailoring. J Clin Endocrinol Metab. 2000;85:159–64. 160. Mastorakos G, Pavlatou M. Exercise as a stress model and the interplay between the hypothalamus-­ pituitary-­ adrenal and the hypothalamus-pituitary-­ thyroid axes. Horm Metab Res. 2005;37:577–84. 161. Uribe RM, Jaimes-Hoy L, Ramírez-Martínez C, García-Vázquez A, Romero F, Cisneros M, Cote-­ Vélez A, Charli JL, Joseph-Bravo P. Voluntary exercise adapts the hypothalamus-pituitary-thyroid axis in male rats. Endocrinology. 2014;155(5):2020–30. 162. Lesmana R, Iwasaki T, Iizuka Y, Amano I, Shimokawa N, Koibuchi N. The change in thyroid hormone sig- 6 Exercise and Thyroid Function 163. 164. 165. 166. 167. 168. 169. 170. 171. 172. 173. 174. 175. 176. 177. 178. naling by altered training intensity in male rat skeletal muscle. Endocr J. 2016;63(8):727–38. Huang WS, Yu MD, Lee MS, et al. Effect of treadmill exercise on circulating thyroid hormone measurements. Med Princ Pract. 2004;13:15–9. Premachandra BN, Winder WW, Hickson R, et al. Circulating reverse triiodothyronine in humans during exercise. Eur J Appl Physiol. 1981;47:281–8. Mason JW, Hartley LH, Kotchen TA, et al. Plasma thyroid stimulating hormone response in anticipation of muscular exercise in the human. J Clin Endocrinol Metab. 1973;37:403–6. Liewendahl K, Helenius T, Niiveri H, et al. Fatty acid-induced increase in serum dialyzable free thyroxine after physical exercise: implication for nonthyroidal illness. J Clin Endocrinol Metab. 1992;74:1361–5. Galbo H, Hummer L, Petersen IB, et al. Thyroid and testicular hormone responses to graded and prolonged exercise in man. Eur J Appl Physiol. 1977;36:101–6. Schmid P, Wolf W, Pilger E, et al. TSH, T3, rT3 and FT4 in maximal and submaximal physical exercise. Eur J Appl Physiol. 1982;48:31–9. Hackney AC, Kallman A, Hosick KP, Rubin DA, Battaglini CL. Thyroid hormonal responses to intensive interval versus steady-state endurance exercise sessions. Hormones (Athens). 2012;11(1):54–60. Hesse V, Vilser C, Scheibe I, et al. Thyroid hormone metabolism under extreme body exercise. Exp Clin Endocrinol. 1989;94:82–8. Semple CG, Thomson LA, Beastall GR. Endocrine responses to marathon running. Br J Sports Med. 1985;19:148–51. Sander M, Rocker L. Influence of marathon running on thyroid hormones. Int J Sports Med. 1988;9:123–6. Limanová Z, Sonka I, Kratochvil O, et al. Effects of exercise on serum cortisol and thyroid hormones. Exp Clin Endocrinol. 1983;81:308–14. Rone IK, Dons RF, Reed HL. The effect of endurance training on serum triiodothyronine kinetics in man: physical conditioning marked by enhanced thyroid hormone metabolism. Clin Endocrinol. 1992;37:325–30. Smallridge RC, Whorton NE, Burman KD, et al. Effects of exercise and physical fitness on the pituitary-­thyroid axis and on prolactin secretion in male runners. Metabolism. 1985;34:949–54. Deligiannis A, Karamouzis M, Kouidi E, et al. Plasma TSH, T3, T4 and cortisol responses to swimming at varying water temperatures. Br J Sports Med. 1993;27:247–50. Dulac S, Quirion A, DeCarufel D, et al. Metabolic and hormonal responses to long-distance swimming in cold water. Int J Sports Med. 1987;8:352–6. Reichlin S, Martin JB, Jackson IMD. Regulation of thyroid stimulating hormone (TSH) secretion. In: Jeffcoate SL, Hutchinson ISM, editors. The endo- 107 179. 180. 181. 182. 183. 184. 185. 186. 187. 188. 189. 190. 191. 192. 193. crine hypothalamus. London: Academic; 1978. p. 237–43. Rhodes BA, Conway MJ. Exercise lowers thyroid radioiodine uptake: concise communication. J Nucl Med. 1980;21:835–7. Tremblay A, Poehlman ET, Despres JP, et al. Endurance training with constant energy intake in identical twins: changes over time in energy expenditure and related hormones. Metabolism. 1997;46:499–503. Lehmann M, Knizia K, Gastmann U, et al. Influence of 6-week, 6 days per week, training on pituitary function in recreational athletes. Br J Sports Med. 1993;27:186–92. Burman KD, Diamond RC, Harvey GS, et al. Glucose modulation of alterations in serum iodothyronine concentrations induced by fasting. Metabolism. 1979;28:291–9. Loucks AB, Heath EM. Induction of low-T3 syndrome in exercising women occurs at a threshold of energy availability. Am J Phys. 1994;264:817–23. Loucks AB, Thuma JR. Luteinizing hormone pulsatility is disrupted at a threshold of energy availability in regularly menstruating women. J Clin Endocrinol Metab. 2003;88(1):297–311. Maughan RJ. Nutrition in sport. Chichester: Wiley; 2008. Ihle R, Loucks AB. Dose-response relationships between energy availability and bone turnover in young exercising women. J Bone Miner Res. 2004;19(8):1231–40. Epub 2004 Apr 19. Mathieson RA, Walberg IT, Gwazdauskas FC, et al. The effect of varying carbohydrate content of a very-­ low-­caloric-diet on resting metabolic rate and thyroid hormones. Metabolism. 1986;35:394–8. O’Connell M, Robbins DC, Horton ES, et al. Changes in serum concentrations of 3,3’,5’triiodothyronine and 3,5,3’-triiodothyronine during prolonged moderate exercise. J Clin Endocrinol Metab. 1979;49:242–6. Opstad PK, Falch D, Okedalen O, et al. The thyroid function in young men during prolonged exercise and the effect of energy and sleep deprivation. Clin Endocrinol. 1984;20:657–9. Hackney AC, Hodgdon JA. Thyroid hormone changes during military field operations: effects of cold exposure in the Arctic. Aviat Space Environ Med. 1992;63(7):606–11. Hackney AC, Feith S, Pozos R, Seale J. Effects of high altitude and cold exposure on resting thyroid hormone concentrations. Aviat Space Environ Med. 1995;66(4):325–9. Sawhney RC, Malhotra AS. Thyroid function in sojourners and acclimatized low landers at high -altitude in man. Horm Metab Res. 1991;23:81. Stock MJ, Chapman C, Stirling JL, Campbell IT. Effects of exercise, altitude, and food on blood -hormone and metabolic levels. J Appl Physiol. 1978;45:350–4. 108 194. Fortunato RS, Ignácio DL, Padron AS, et al. The effect of acute exercise session on thyroid hormone economy in rats. J Endocrinol. 2008;198: 347–53. 195. Simsch C, Lormes W, Petersen KG, et al. Training intensity influences leptin and thyroid hormones in highly trained rowers. Int J Sports Med. 2002;23:422–7. 196. Benso A, Broglio F, Aimaretti G, et al. Endocrine and metabolic responses to extreme altitude and D. Ylli et al. physical exercise in climbers. Eur J Endocrinol. 2007;157:733–40. Soc Ital Biol Sper. 1984;60:753–9. 197. Harber VJ, Petersen SR, Chilibeck PD. Thyroid hormone concentrations and muscle metabolism in amenorrheic and eumenorrheic athletes. Can J Appl Physiol. 1998;23:293–306. 198. Creatsas G, Salakos N, Averkiou M, et al. Endocrinological profile of oligomenorrheic strenuously exercising adolescents. Int J Gynaecol Obstet. 1992;38:215–21. 7 The Male Reproductive System, Exercise, and Training: Endocrine Adaptations Fabio Lanfranco and Marco Alessandro Minetto Introduction Androgens exert strong anabolic effects on skeletal muscle protein synthesis [1, 2], satellite cell number [3], and skeletal muscle growth [4, 5]. Because these changes are of great importance to muscle mass and strength, androgens have been recognized as important hormones that influence sports performance [6]. Exerciseinduced changes in testosterone concentrations can moderate or support neuromuscular performance through various short-term mechanisms (e.g., second messengers, lipid/protein pathways, neuronal activity, behavior, cognition, motor system function, muscle properties, and energy metabolism) [7]. On the other hand, the gonadal axis function is strongly affected by physical exercise depending on the intensity and duration of the activity, the fitness level, and the nutritionalmetabolic status of the individual [8–10]. Moreover, circulating testosterone and its bioavailable fractions are affected by weight and F. Lanfranco (*) AOU Citta della Salute e della Scienza di Torino, Division of Endocrinology, Diabetology and Metabolism, University of Turin, Department of Medical Sciences, Turin, Italy e-mail: fabio.lanfranco@unito.it M. A. Minetto Division of Physical Medicine and Rehabilitation, Department of Surgical Sciences, University of Turin, Turin, Italy age. They are also changed by different kinds of stress which may appear as physical stress (i.e., endurance training, sleep deprivation in extreme sports, changes of air pressure in altitude training) or mental stress in relation to sport events and training [9, 10]. The purpose of this chapter is to illustrate the physiologic and pathologic changes that occur in the male gonadal axis secondary to acute exercise and chronic exercise training. hysiology of the Male Gonadal P Axis The male gonadal axis consists of the testes and the hypothalamus-pituitary unit that controls their function. The testes possess a dual function, i.e., the production of androgens and of the sperm. Figure 7.1 depicts an outline of the male gonadal axis and of the hormonal regulation of the testicular function. The pituitary gland is the central structure controlling gonadal function: it releases the gonadotropins luteinizing hormone (LH) and follicle-stimulating hormone (FSH) and is regulated by the hypothalamic gonadotropin-­ releasing hormone (GnRH), which is secreted in a pulsatile fashion with peaks every 90–120 min. GnRH secretion is modulated by a network of excitatory and inhibitory inputs that include either a central control exerted by distinct © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_7 109 F. Lanfranco and M. A. Minetto 110 Cortex DA Stress Nutrition Exercise – PRL – Hypothalamus GnRH – + – – + – – + GABA – NPY Leptin IL-1 – + NE CRH Cortisol Serotonin β-endorphins + Ghrelin – LH FSH + + Ghrelin CRH Cortisol – + Inhibins Activins – – Estradiol Testosterone Leydig cell DHT Sertoli cell Sperm – ROS Fig. 7.1 Schematic diagram of the male gonadal axis. CRH corticotropin-releasing hormone, DA dopamine, DHT dihydrotestosterone, FSH follicle-stimulating hormone, GABA gamma-aminobutyric acid, GnRH gonadotropin-­releasing hormone, IL-1 interleukin-1, LH luteinizing hormone, NE norepinephrine, NPY neuropeptide Y, PRL prolactin, ROS reactive oxygen species subgroups of neurons afferent to the GnRHsecreting neurons or the peripheral gonadal steroid feedback [11]. The hypothalamic kisspeptin system exerts a fundamental control in the activation of GnRH-­ secreting neurons at puberty [11]. In addition, several neurotransmitters and neuromodulators are proposed to influence GnRH secretion: the noradrenergic system and neuropeptide Y (NPY) show stimulatory activity, whereas interleukin-1, opioid peptides, dopamine, serotonine, and gamma-aminobutyric acid (GABA) are inhibitory. Another important peptide regulating the GnRH secretion is leptin, a peptide hormone secreted by the adipose tissue that helps to regulate the energy balance and mirrors the amount of energy reserve. Leptin purportedly plays a key role either by signaling to the central nervous system the information regarding the amount of available fat stores or by enabling the activation of the gonadal axis through the GnRH secretion 7 The Male Reproductive System, Exercise, and Training: Endocrine Adaptations when convenient [11]. Specifically, leptin has been shown to stimulate GnRH and gonadotropin secretions [12]. Additionally, ghrelin, a peptide hormone with growth hormone-releasing action, exerts multiple endocrine and non-endocrine effects including inhibition of the gonadal axis at both the central and peripheral level [13]. Another important mechanism that dynamically controls GnRH synthesis and release is represented by the gonadal steroid feedback. In man, the major hormone controlling GnRH secretion is testosterone, which inhibits gonadotropin secretion via negative feedback both at the hypothalamic and pituitary level. Dihydrotestosterone (DHT) and estradiol also modulate gonadotropin secretion acting at the hypothalamic and/or pituitary level [11, 12, 14, 15]. Noteworthy, the kisspeptin system is implicated in the transmission of both negative and positive feedback of sex steroids on GnRH neurons [11]. Finally, the adverse effect of stress on reproductive function is well known. Several hormonal factors are involved: corticotropin-releasing hormone (CRH) inhibits GnRH secretion, prolactin (PRL) further reduces the GnRH pulse rate [14], and cortisol inhibits both the hypothalamus-­ pituitary and gonadal functions. LH and FSH are produced and secreted by the gonadotropic cells of the anterior pituitary. LH regulates testicular androgenesis, whereas FSH, together with locally produced testosterone, is responsible for spermatogenesis. LH binds to specific receptors on the surface of Leydig cells in the testis and regulates the biosynthesis of testosterone. FSH binds to receptors on the Sertoli cells and promotes spermatogenesis: in addition to a number of other proteins, the hormones inhibin B and activins are formed in the Sertoli cells under the influence of FSH. Inhibin B plays an important role in the feedback regulation of FSH secretion, whereas the physiological role of activins has not been conclusively clarified thus far [14, 16]. Testosterone is the most important steroid produced by the testis and is responsible for the development and maintenance of male sex characteristics as well as a number of other anabolic and metabolic effects (e.g., muscle and 111 bone metabolism). Testosterone is produced primarily in the Leydig cells of the testis. It may be further metabolized into a more potent androgen, DHT. Normal testosterone concentrations in adult males range between 12 and 30 nmol/l: testosterone concentrations in blood follow a circadian rhythm with higher levels in the morning hours and about 25% lower levels in the evening [12, 17]. ffects of Physical Exercise E on Testicular Steroidogenesis hort, Intense Exercise Increases S Circulating Testosterone The effects of physical activity on the male gonadal axis vary with the intensity and duration of the activity, the fitness level of the individual, and his nutritional-metabolic status. Relatively short, intense exercise usually increases, while more prolonged exercise usually decreases serum testosterone levels [8, 9, 18, 19]. Increased blood testosterone levels have been reported during relatively strenuous free and treadmill running, weight training, rock climbing, and ergometer cycling [20–22]. Shortterm sprints can be seen as strength outburst and are comparable to resistance exercise rather than endurance exercise: in fact, sprint exercise increased blood testosterone concentrations in adolescent boys [23]. The testosterone response increases with increased exercise load [24]. Similar workloads produce similar responses, regardless of whether the load is aerobic or anaerobic [25]. Immediate and 5-min post-exercise measurements showed an increase in testosterone levels both in men and women [26]. Acute exercise-­ induced testosterone increments are also seen in older men [27]. This acute hormone response was confirmed and described to be markedly stronger in young men compared to old in a study involving ten men with mean age 26.5 years and ten men with mean age 70.0 years [28]. As muscle mass increases with strength training [4] and is correlated with testosterone 112 levels, it could be expected that the testosterone response to acute exercise is higher in persons constantly involved in strength training. Consistently, a 6-month sprint training program increased plasma testosterone concentrations in response to sprint exercise in adolescent boys [23]. Experienced weight lifters compared to beginners showed similar basal levels of testosterone but were able to evoke a stronger testosterone response during exercise [20]. Contrary to these findings, a long-term training period of 12 weeks involving younger (mean 23 years) and older men (mean 63 years) showed no significant results concerning testosterone levels before or immediately after exercise [29]. Ronnestad et al. [30] investigated the effects of testosterone and growth hormone (GH) transient increase during exercise, indicating that performing leg exercises prior to arm exercises, thereby increasing the levels of testosterone and GH, induced superior strength training adaptations compared to arm training without acute elevation of hormones. It has been found that acute elevation in endogenous testosterone (by strength training) potentiates the androgen receptor (AR) response to a strength training session compared to no acute elevation of endogenous testosterone [31]. It might thus be speculated that the results by Ronnestad et al. are due to an increased AR expression, and through an improved testosterone-receptor interaction, and a subsequent increased protein synthesis, leading to superior strength training adaptations. This hypothesis has also been evaluated by Ahtiainen et al. [32], who have described a correlation of individual pre- to post-training changes in resting AR protein concentration with the changes in cross-sectional area of muscle fibers in a combined group of young and elderly subjects who performed heavy resistance exercise bouts before and after a training period. Collectively, these findings suggested that the individual changes of AR protein concentration in skeletal muscle following resistance training may have a critical impact on training-induced muscular adaptations. F. Lanfranco and M. A. Minetto echanisms Underlying Increases M in Circulating Testosterone Following Short, Intense Exercise No conclusive and homogeneous evidence about gonadotropin response to an acute exercise bout is available. In fact, LH and FSH levels have been reported to be increased, decreased, or unchanged by short-term strenuous exercise [33–36]. The exercise-associated increment in circulating testosterone is considered not to be mediated by LH, due to the inconsistent LH response and to the evidence that testosterone levels increase more quickly than LH in response to exercise. Possible mechanisms such as hemoconcentration, reduced clearance, and/or increased testosterone synthesis may be involved [34, 36–38]. However, the timing of testosterone response differs from that of other circulating steroids (e.g., androstenedione and dehydroepiandrosterone increase simultaneously with cortisol), thus suggesting that specific testicular mechanisms are involved [36]. These mechanisms may include the activation of the sympathetic system, which stimulates testicular testosterone production during exercise via a direct neural pathway in some species [39]. Catecholamine levels also increase significantly during exercise. Beta-adrenergic blockade inhibits testosterone responses to exercise, whereas L-dopa, phentolamine, and clonidine had no effect [40]. An anticipatory increase in circulating testosterone levels has also been described and seems to be independent of hepatic perfusion or hemoconcentration [33, 36]. Ultimately, the exact mechanisms involved in increasing testosterone concentrations in specific exercise protocols are yet to be delineated. rolonged, Submaximal Exercise P and Chronic Exercise Training Decrease Circulating Testosterone: From the “Female Athlete Triad” to “Relative Energy Deficiency in Sport (RED-S)” In contrast to the short-term testosterone increment during and immediately after short, intense 7 The Male Reproductive System, Exercise, and Training: Endocrine Adaptations 113 exercise, a suppression of serum testosterone lev- rather seen between the so-called high and low els occurs during and subsequent to prolonged responders. Each group has a specific endocrine exercise, in the hours following intense exercise, reactivity pattern concerning the hypothalamus-­ as well as during chronic exercise training [10]. pituitary-­ adrenal axis [44]. It seems that the During the last decades, an increasing number decrease of testosterone levels under the stressful of investigative research studies have pointed to situations of endurance sport is not sufficiently how chronic exposure to endurance exercise answered by the pituitary. There is no adequate training can result in the development of a dys- rise in LH levels, which seem to be unaltered or function within the reproductive components of even show a tendency to decrease with the growthe neuroendocrine system. The majority of these ing amount of stress impact. Nevertheless, age-­ studies have concentrated upon women. However, dependent effects seem to exist in this regard, and the effects of endurance exercise training on the the ratio of androgen to estradiol is shifted by male reproductive neuroendocrine system have physical activity to a more favorable pattern been investigated beginning in the 1980s [41]. (higher androgen and lower estradiol levels) in Most studies observed athletes during training older men compared to younger men performing and competition, giving the impression of gener- regular mild physical activity [45]. ally lowered androgen levels, but lack the comParticipation in sports where leanness is conparison with a control group [9]. sidered a competitive advantage, such as running, A controlled study examining the effects of cycling, wrestling, lightweight rowing, and gymendurance training on the hypothalamus-­ nastics, has been linked to lower body mass index pituitary-­testis axis involved 53 men undergoing (BMI) [46], eating disorders [47], and low energy endurance training for at least 5 years and a con- availability [48]. Low energy availability in the trol group of 35 age-matched, sedentary men. context of anorexia nervosa has been associated Baseline serum testosterone levels of the exer- with low testosterone levels in males [49]. cising men were significantly lower than in the Hagmar et al. [50] evaluated athletes from 26 difcontrol group. Differences in gonadotropins ferent sports and divided them into those who were not seen. Normal regulation would require participated in leanness sports and those who did LH levels to rise with falling testosterone levels, not. The leanness sport athletes had lower body as these have a positive feedback on pituitary fat, higher spinal bone mineral density (BMD), gonadotropin release. A suppression in the regu- lower serum-free testosterone and leptin, and latory axis has been proposed as an explanation higher IGF-1 binding protein. The authors sugof this finding [42]. gested that the increase in BMD could be because Contrary to these observations, basal testos- of the increase in mechanical loading in the speterone levels in trained weight lifters were not cific leanness sports, which presumably overaltered, nor did an increase in the daily training came the effects of lower testosterone and leptin, volume change these levels [43]. Similarly, basal both of which are bone anabolic hormones. testosterone, free testosterone, bioavailable tesFagerberg [51] has recently outlined the negatosterone, and sex hormone-binding globulin tive consequences of low energy availability in concentrations were not significantly different in male bodybuilding. Bodybuilding is a sport in high top-class athletes (sprinters and jumpers) vs. which athletes compete to show muscular definiuntrained subjects [22]. tion, symmetry, and low body fat. The process of Endurance training can be seen as a factor of contest preparation in bodybuilding includes exposure not only to physical but also to psycho- months of underfeeding, thus increasing the risk logical stress. It has been demonstrated in a con- of low energy availability and its negative health trolled study that the reactivity patterns of mental/ consequences, including extreme effects on cirpsychological and physical stress response of the culating testosterone levels. hypothalamus-pituitary-adrenal axis are the same In female athletes, low energy availability is in a specific individual. Differential reactivity is a component of the female athlete triad, a term 114 used to describe the interrelationship of decreased energy availability, subsequent hypothalamus-­pituitary-­gonadal axis inhibition leading to menstrual irregularity, and decreased bone mineral density [48]. The triad was first described by the American College of Sports Medicine (ACSM) in the 1990s. In 2007, the ACSM published a revised position stand on the female athlete triad describing it more broadly as the harmful effects of low energy availability on menstrual function and bone mineral density [52]. The International Olympic Committee (IOC) has recently proposed an expansion of the concept of the female athlete triad to include males and has coined the term “relative energy deficiency in sport (RED-S)” [53]. The development of the term RED-S had three main purposes: (1) to draw awareness to the fact that energy restriction can have negative consequences in men in addition to women; (2) to highlight other potential negative health and performance consequences of low energy availability in athletes besides bone problems; and (3) to encourage expansive research into the potential myriad effects of low energy availability in various populations, including paralympic athletes [10]. The “Exercise-Hypogonadal Male Condition”: Clinical Issues It has been demonstrated that among subjects engaged in chronic exercise training, a selected group of men develop alterations in their reproductive hormonal profile, i.e., persistently low basal resting testosterone concentrations [54, 55]. In particular, the majority of these men exhibit clinically “normal” testosterone concentrations, but these concentrations are at the low end of normal range or even reach subclinical status. In 2005, Hackney and associates proposed the use of “the exercise-hypogonadal male” as a label for this condition [56]. The health consequences of such hormonal changes are increased risk of abnormal spermatogenesis, male infertility problems, and compromised bone mineralization [54, 55, 57, 58]. F. Lanfranco and M. A. Minetto Without large-scale epidemiological studies in this area, clear prevalence data is not available [59]. However, several studies show a clear and consistently reduced serum testosterone concentration in highly aerobically trained individuals, suggesting the exercise-hypogonadal male condition (EHMC) can be a common response [42, 59, 60]. It also appears that as the level of athlete increases, so too does the incidence and severity of the condition [61]. In addition, with no long-­ term data currently available, it is unclear whether the presence of reduced testosterone varies throughout a competitive season and how long it takes for testosterone to return to normal, if at all [59]. The time course for the development of the EHMC or the threshold of exercise training necessary to induce the condition remains unresolved, but preliminary evidence suggest an extended window of time (i.e., years) may be necessary for its development [55] . EHMC shares similarities with overreaching or overtraining and has also been described in male athletes as a parallel process to the female athlete triad, with hypogonadism replacing functional hypothalamic amenorrhea [62, 63]. The existence of the EHMC fits into the terminology of RED-S as clinical manifestation of it may include sexual dysfunction like infertility and reduced libido as well as reduced BMD with associated increase in risk of bone stress injury [59]. The “Exercise-Hypogonadal Male Condition”: Pathophysiological Mechanisms Exercise-hypogonadal men frequently display a lack of significant elevation in basal LH in correspondence with the reduced testosterone concentration, reflecting hypogonadotrophichypogonadism characteristics [41, 54, 64]. These LH abnormalities may involve disparities in luteinizing pulsatility (i.e., pulse frequency and amplitude), although evidence for altered LH pulsatile release is conflicting [65, 66]. Moreover, gonadotropin response to 7 The Male Reproductive System, Exercise, and Training: Endocrine Adaptations GnRH has been reported both reduced and increased following prolonged, exhaustive exercise [67, 68]. Exercise-hypogonadal men have been shown to have altered basal PRL [54]. At either excessively low or high circulating levels, PRL can result in suppression of testosterone levels in men [69]. It has been speculated that the absence of PRL at the testicle alters the effectiveness of LH to stimulate testosterone production. This theory is based upon the proposed synergistic effects of PRL upon testicular LH receptors [41]. However, not all investigators reporting low resting testosterone in endurance-trained men have reported the concomitant existence of low resting PRL levels [69]. Some investigations have looked at a potential relationship between high PRL levels and low testosterone, speculating that any “stressful” situation might provoke disproportionate PRL responses in exercise-hypogonadal men and this ultimately promotes a reproductive axis disruption [70]. As previously mentioned, leptin and ghrelin are two hormones associated with appetite regulation which function as metabolic modulators of the gonadotropic axis, as well [13, 71]. Acute and chronic exercise can impact upon resting leptin and ghrelin concentrations, independent of changes in body adiposity [72, 73]. However, to date no research studies have examined whether leptin and/or ghrelin concentrations are altered in exercise-hypogonadal men. Such work would be illuminating on the topic and is needed. Other research investigations have focused on alterations in testicular ability to produce and secrete testosterone and to respond to exogenous stimuli (i.e., LH or hCG). Whereas animal studies have demonstrated that exercise training compromises testicular enzymatic activity [74], data in exercise-hypogonadal men are contradictory. In fact, some investigations suggest testicular steroidogenesis is normal, while some indicate it is marginally impaired when challenged with exogenous stimuli [54]. Another potential disruptive hormone to the gonadal axis is cortisol. Studies in a wide range of sports (e.g., cycling, marathon running, football, handball, rugby, tennis, swimming, and 115 wrestling) have almost all shown increased cortisol concentrations during exercise [75, 76]. Cortisol secretion increases in response to exercise intensity and duration, as well as to the training level of subjects [77–80], at least in part to mobilize energy stores. An inhibitory effect of the hypothalamus-pituitary-adrenal axis on the reproductive system has been demonstrated in both sexes [81, 82]. In fact, glucocorticoids suppress gonadal axis function at the hypothalamic-­ pituitary level [81]. Moreover, Inder et al. [83] have demonstrated that dexamethasone administration in humans reduces circulating testosterone and downregulates the muscular expression of the AR. Finally, CRH and its receptors have been identified in the Leydig cells of the testis, where CRH exerts inhibitory actions on testosterone biosynthesis [84]. Interestingly, a sporting event and also training for such represent both a physical and a mental stress [9]. The release of cortisol by activation of the hypothalamic-pituitary-adrenal axis as reaction to mental stress is well documented, especially in competitive situations [44, 85]. Stress responses by the hypothalamic-pituitary-­ gonadal axis are constantly found as well. Along this line, anticipatory stress was measured in 50 males before a 1-day experimental stress event (participation in stressful clinical research protocol). Cortisol levels rose significantly, while both testosterone and LH secretion were decreased [86]. Psychological stress markers as measured by scales for anxiety, hostility, and depression were correlated with serum levels of testosterone in a group of males aged 30–55 years. Those classified as highly stressed had significantly lower testosterone levels than their counterparts [87]. A cross-sectional study involving 439 males all aged 51 years showed those with low levels of testosterone (adjusted for body mass index) to exhibit a cluster of psychosocial stress indicators [88]. Nevertheless, other hormonal profile studies reporting the existence of low testosterone in trained men did not show elevated resting cortisol levels suggesting that the hypothalamicpituitary-adrenal axis is not playing any role in the development of EHMC [41, 59, 60, 89]. 116 However, resting cortisol levels do not necessarily reflect a hyperactivity of the hypothalamus-pituitary-adrenal axis, which can be better defined either by serial blood or salivary sampling [90] or by assay of urinary free cortisol. Thus, at this time the role of cortisol to the changes found in the gonadal axis of trained men is in need of further studies. ffects of Physical Exercise E on Spermatogenesis Clinical expression of impaired reproductive function in men engaged in chronic exercise training seems uncommon [57, 66, 91]. However, chronic physical exercise may induce a state of oligospermia, a reduction of the total number of motile sperm and an increase in abnormal or immature spermatozoa. Increase in “round cells” has also been reported indicating a possible infectious and/or inflammatory environment [57]. Arce and colleagues [57] were able to retrospectively establish an exercise (i.e., running) volume threshold of 100 km/wk for semen alterations to occur, as they found alterations in sperm density, motility, morphology, and in vitro sperm penetration of standard cervical mucus in endurance-­ trained runners when compared to resistance athletes or sedentary subjects. Similarly, Safarinejad et al. [68] observed a negative effect of training on sperm parameters in high-intensity training athletes when compared to moderate-intensity ones. Scientific evidence seems to support the existence of a minimum level of volume for detrimental effects to take place, either hormonal or seminological [54, 57]. As Hackney et al. [54, 56] highlight, alterations may well represent the accumulative effect, more than the acute response, of years of training load. Some of the latest research has shown that training intensity, and not only volume, is greatly important in this equation as well. In fact, Vaamonde et al. [92] point out that sperm DNA damage and alteration are oxidative stress-related parameters. F. Lanfranco and M. A. Minetto High-level athletes have been typically training for many years, making it difficult to establish a potential harmful training threshold (volume and/or intensity) as they normally start training at pre- or peri-pubertal years [93]. Nevertheless, high volume cycling training seems to correlate with sperm morphology anomalies. Wise et al. [94] have examined the association between regular physical activity and semen quality in a large cohort of 2261 men attending an infertility clinic. They found that none of the semen parameters (semen volume, sperm concentration, sperm motility, sperm morphology, and total motile sperm) were materially associated with regular exercise. However, in the subgroup of men who reported bicycling as their primary form of exercise, bicycling at levels of >5 h/wk was associated with low sperm concentration and total motile sperm. These findings generally agree with earlier studies that have shown deleterious effects of bicycling on semen parameters among competitive cyclists [91, 95]. It remains unclear as to whether the changes associated with bicycling are due to mechanical trauma (i.e., caused by compression of scrotum on the bicycle saddle), to a prolonged increase in core scrotal temperature (i.e., related to exercise itself or wearing of constrictive clothing), or some other factors [96]. xidative Stress as a Putative O Mechanism Underlying Impaired Spermatogenesis in Exercise-­ Hypogonadal Men Several mechanisms have been reported to affect the male reproductive function in exercising subjects. Alterations in the hormonal milieu, as discussed in the previous paragraph, may well play a role, since qualitatively and quantitatively normal spermatogenesis is critically dependent on an intact hypothalamus-pituitarytestis axis. On the other hand, it has been reported that endurance exercise is associated with oxidative stress [97]. During endurance exercise, there is a 10- to 20-fold increase in 7 The Male Reproductive System, Exercise, and Training: Endocrine Adaptations whole body oxygen (O2) consumption, and O2 uptake in the active skeletal muscle increases 100- to 200-fold [98]. This increase in O2 utilization may result in the production of reactive oxygen species (ROS) at the rates that exceed the body’s capacity to detoxify them [99]. An increase in the formation of ROS decreases fertility, as the ROS will attack the membranes of the spermatozoa, decreasing their viability [100]. Vaamonde et al. [101] have reported exercise-related alterations in sperm which may be prevented with antioxidant agents. Vaamonde et al. [102] also reported that, similarly to sperm morphology, cycling volume positively correlates to sperm DNA fragmentation, also observing high correlation between training volume, sperm DNA fragmentation, and percentage of morphological abnormalities [93]. However, an increasing number of studies suggest that exercise training enhances antioxidant capacity [103, 104]. Indeed, the machinery eliminating ROS adapts after regular exercise and actually lowers the amount of ROS that is produced, especially in the major organs (muscles) of oxygen consumption and ROS production. In recent years, the anti-inflammatory and antioxidant properties of regular exercise training have prompted some investigators to explore the effects of different exercise modalities on markers of inflammation and oxidative stress in seminal plasma [105, 106]. Hajizadeh Maleki and colleagues have conducted independent randomized controlled trials looking at the effects of exercise training at different intensity levels on markers of reproductive function and reproductive performance in infertile and fertile men and demonstrated significant improvements in a variety of sperm oxidative stress and inflammation assays as well as semen quality and sperm DNA integrity following 24 weeks of exercise training, suggesting that regular resistance exercise, in particular at a moderate intensity level, positively affects the markers of male reproduction [105, 107–109]. However, how changes in seminal markers of male reproductive function may be connected with reproductive outcomes remains to be determined. 117 Conclusions The male gonadal axis function is strongly affected by physical exercise. Relatively short, intense exercise usually increases, while more prolonged exercise usually decreases serum testosterone levels. Restricted energy availability may negatively affect hormone levels both in female and in male endurance athletes as highlighted by the definition of “relative energy deficiency in sport (RED-S)” by the IOC. Reduced or low-normal circulating testosterone concentrations involve health consequences such as an increased risk of abnormal spermatogenesis, infertility problems, and compromised bone mineralization. Thus, awareness must be raised that exercise can represent a potential cause of andrological problems. On the other hand, moderate and low-level exercise has been recently shown to exert positive effects on the male reproductive potential. Ultimately, additional research is needed in this area with proper standardization in assessment tools and study protocols to draw more accurate conclusions about the effects of physical exercise on the male gonadal axis function. References 1. Urban RJ, Bodenburg YH, Gilkison C, et al. Testosterone administration to elderly men increases skeletal muscle strength and protein synthesis. Am J Phys. 1995;269:E820–6. 2. Ferrando AA, Sheffield-Moore M, Yeckel CW, et al. Testosterone administration to older men improves muscle function: molecular and physiological mechanisms. Am J Physiol Endocrinol Metab. 2002;282:E601–7. 3. Sinha-Hikim I, Roth SM, Lee MI, Bhasin S. Testosterone-induced muscle hypertrophy is associated with an increase in satellite cell number in healthy, young men. Am J Physiol Endocrinol Metab. 2003;285:E197–205. 4. Bhasin S, Storer TW, Berman N, et al. The effects of supraphysiologic doses of testosterone on muscle size and strength in normal men. N Engl J Med. 1996;335:1–7. 5. Sinha-Hikim I, Artaza J, Woodhouse L, et al. Testosterone-induced increase in muscle size in healthy young men is associated with muscle fiber F. Lanfranco and M. A. Minetto 118 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. hypertrophy. Am J Physiol Endocrinol Metab. 2002;283:E154–64. Viru A, Viru M. Preconditioning of the performance in power events by endogenous testosterone: in memory of professor Carmelo Bosco. J Strength Cond Res. 2005;19:6–8. Crewther BT, Cook C, Cardinale M, Weatherby RP, Lowe T. Two emerging concepts for elite athletes: the short-term effects of testosterone and cortisol on the neuromuscular system and the dose-response training role of these endogenous hormones. Sports Med. 2011;41:103–23. Vingren JL, Kraemer WJ, Ratamess NA, Anderson JM, Volek JS, Maresh CM. Testosterone physiology in resistance exercise and training: the up-stream regulatory elements. Sports Med. 2010;40:1037–53. Zitzmann M. Exercise, training, and the hypothalamic-­ pituitary-gonadal axis in men. Hormone use and abuse Athletes, E. Ghigo, F. Lanfranco, C.J. Strasburger, Springer New York, 2011, 29,: https://doi.org/10.1007/978-1-44197014-5, pp 25–30. Cano Sokoloff N, Misra M, Ackerman KE. Exercise, training, and the hypothalamic-pituitary-gonadal axis in men and women. Front Horm Res. 2016;47:27–43. Bonomi M, Vezzoli V, Cariboni A. Control of GnRH secretion. In: Simoni M, Huhtaniemi IT, editors. Endocrinology of the testis and male reproduction, Endocrinology 1. Switzerland: Springer International Publishing AG; 2017. p. 3–33. Weinbauer GF, Luetjens CM, Simoni M, Nieschlag E. Physiology of testicular function. In: Nieschlag E, Behre H, Nieschlag S, editors. Andrology: male reproductive health and dysfunction. 3rd ed. Heidelberg: Springer; 2009. p. 11–60. Lanfranco F, Bonelli L, Baldi M, et al. Acylated ghrelin inhibits spontaneous LH pulsatility and responsiveness to naloxone, but not that to GnRH in young men: evidence for a central inhibitory action of ghrelin on the gonadal axis. J Clin Endocrinol Metab. 2008;93:3633–9. Jockenhoevel F, Schubert M. Anatomy and physiology of the testis. In: Jockenhoevel F, Schubert M, editors. Male hypogonadism. Bremen: UNI-MED Verlag; 2007. p. 12–30. Herbison AE. Control of puberty onset and fertility by gonadotropin-releasing hormone neurons. Nat Rev Endocrinol. 2016;12:452–66. Ulloa-Aguirre A, Dias JA, Bousfield GR. Gonadotropins. In: Simoni M, Huhtaniemi IT, editors. Endocrinology of the testis and male reproduction, Endocrinology 1. Switzerland: Springer International Publishing AG; 2017. p. 71–122. Flueck CE, Pandey AV. Testicular steroidogenesis. In: Simoni M, Huhtaniemi IT, editors. Endocrinology of the testis and male reproduction, Endocrinology 1. Switzerland: Springer International Publishing AG; 2017. p. 343–71. 18. Cumming DC, Wheeler GD, McColl EM. The effects of exercise on reproductive function in men. Sports Med. 1989;7:1–17. 19. Di Luigi L, Romanelli F, Sgrò P, Lenzi A. Andrological aspects of physical exercise and sport medicine. Endocrine. 2012;42:278–84. 20. Kraemer RR, Kilgore JL, Kraemer GR, Castracane VD. Growth hormone, IGF-I, and testosterone responses to resistive exercise. Med Sci Sports Exerc. 1992;24:1346-1352. 21. Sherk VD, Sherk KA, Kim S, Young KC, Bemben DA. Hormone responses to a continuous bout of rock climbing in men. Eur J Appl Physiol. 2011;111:687–93. 22. Grandys M, Majerczak J, Zapart-Bukowska J, Kulpa J, Zoladz JA. Gonadal hormone status in highly trained sprinters and in untrained men. J Strength Cond Res. 2011;25:1079–84. 23. Derbré F, Vincent S, Maitel B, et al. Androgen responses to sprint exercise in young men. Int J Sports Med. 2010;31:291–7. 24. Gotshalk LA, Loebel CC, Nindl BC, et al. Hormonal responses of multiset versus single-set heavy-­ resistance exercise protocols. Can J Appl Physiol. 1997;22:244–55. 25. Hackney AC, Premo MC, McMurray RG. Influence of aerobic versus anaerobic exercise on the relationship between reproductive hormones in men. J Sports Sci. 1995;13:305–11. 26. Kraemer WJ, Staron RS, Hagerman FC, et al. The effects of short-term resistance training on endocrine function in men and women. Eur J Appl Physiol. 1998;78:69–76. 27. Häkkinen K, Pakarinen A. Acute hormonal responses to heavy resistance exercise in men and women at different ages. Int J Sports Med. 1995;16:507–13. 28. Häkkinen K, Pakarinen A, Newton RU, Kraemer WJ. Acute hormone responses to heavy resistance lower and upper extremity exercise in young versus old men. Eur J Appl Physiol. 1998;77:312–9. 29. Craig BW, Brown R, Everhart J. Effects of progressive resistance training on growth hormone and testosterone levels in young and elderly subjects. Mech Ageing Dev. 1989;49:159–69. 30. Ronnestad BR, Nygaard H, Raastad T. Physiological elevation of endogenous hormones results in superior strength training adaptation. Eur J Appl Physiol. 2011;111:2249–59. 31. Spiering BA, Kraemer WJ, Vingren JL, et al. Elevated endogenous testosterone concentrations potentiate muscle androgen receptor responses to resistance exercise. J Steroid Biochem Mol Biol. 2009;114:195–9. 32. Ahtiainen JP, Hulmi JJ, Kraemer WJ, et al. Heavy resistance exercise training and skeletal muscle androgen receptor expression in younger and older men. Steroids. 2011;76:183–92. 33. Wilkerson JE, Horvath SM, Gutin B. Plasma testosterone during treadmill exercise. J Appl Physiol. 1980;49:249–53. 7 The Male Reproductive System, Exercise, and Training: Endocrine Adaptations 34. Metivier G, Gauthier R, de la Cevrotriere J, Grymala D. The effect of acute exercise on the serum levels of testosterone and luteinizing (LH) hormone in human male athletes. J Sports Med Phys Fitness. 1980;20:235–7. 35. Schmid P, Pusch PP, Wolf WW, et al. Serum FSH, LH and testosterone in humans after physical exercise. Int J Sports Med. 1982;3:84–9. 36. Cumming DC, Brunsting LA 3rd, Strich G, et al. Reproductive hormone increases in response to acute exercise in men. Med Sci Sports Exerc. 1986;18:369-373. 37. Sutton JR, Coleman MJ, Casey J, Lazarus L. Androgen responses during physical exercise. Br Med J. 1973;1:520–2. 38. Cadoux-Hudson TA, Few JD, Imms FJ. The effect of exercise on the production and clearance of testosterone in well trained young men. Eur J Appl Physiol Occup Physiol. 1985;54:321–5. 39. Levin J, Lloyd CW, Lobotsky J, Friedrich EH. The effect of epinephrine on testosterone production. Acta Endocrinol. 1967;55:184–92. 40. Jezová D, Vigas M. Testosterone response to exercise during blockade and stimulation of adrenergic receptors in man. Horm Res. 1981;15:141–7. 41. Wheeler GD, Wall SR, Belcastro AN, Cumming DC. Reduced serum testosterone and prolactin levels in male distance runners. JAMA. 1984;27:514–6. 42. Hackney AC, Fahrner CL, Gulledge TP. Basal reproductive hormonal profiles are altered in endurance trained men. J Sports Med Phys Fitness. 1998;38:138–41. 43. Fry AC, Kraemer WJ, Ramsey LT. Pituitary-adrenal-­ gonadal responses to high-intensity resistance exercise overtraining. J Appl Physiol. 1998;85:2352–9. 44. Singh A, Petrides JS, Gold PW, Chrousos GP, Deuster PA. Differential hypothalamicpituitary-­ adrenal axis reactivity to psychological and physical stress. J Clin Endocrinol Metab. 1999;84:1944–8. 45. Slowinska-Lisowska M, Jozkow P, Medras M. Associations between physical activity and the androgenic/estrogenic status of men. Physiol Res. 2010;59:757–63. 46. Hagmar M, Hirschberg AL, Berglund L, Berglund B. Special attention to the weight-control strategies employed by Olympic athletes striving for leanness is required. Clin J Sport Med. 2008;18:5–9. 47. Martinsen M, Bratland-Sanda S, Eriksson AK, Sundgot-Borgen J. Dieting to win or to be thin? A study of dieting and disordered eating among adolescent elite athletes and non-athlete controls. Br J Sports Med. 2010;44:70–6. 48. De Souza MJ, Nattiv A, Joy E, et al. Female athlete triad coalition consensus statement on treatment and return to play of the female athlete triad: 1st international conference held in San Francisco, California, May 2012 and 2nd international conference held in Indianapolis, Indiana, May 2013. Br J Sports Med. 2014;48:289. 119 49. Misra M, Katzman DK, Cord J, et al. Bone metabolism in adolescent boys with anorexia nervosa. J Clin Endocrinol Metab. 2008;93:3029–36. 50. Hagmar M, Berglund B, Brismar K, Hirshberg AL. Body composition and endocrine profile of male Olympic athletes striving for leanness. Clin J Sport Med. 2013;23:197–201. 51. Fagerberg P. Negative consequences of low energy availability in natural male bodybuilding: a review. Int J Sport Nutr Exerc Metab. 2018;28:385–402. 52. Nattiv A, Loucks AB, Manore MM, Sanborn CF, Sundgot-Borgen J, Warren MP. American College of Sports Medicine. American College of Sports Medicine position stand. The female athlete triad. Med Sci Sports Exerc. 2007;39:1867–82. 53. Mountjoy M, Sundgot-Borgen J, Burke L, et al. The IOC consensus statement: beyond the female athlete triad--relative energy deficiency in sport (RED-S). Br J Sports Med. 2014;48:491–7. 54. Hackney AC. Effects of endurance exercise on the reproductive system of men: the “exercise-­ hypogonadal male condition”. J Endocrinol Investig. 2008;31:932–8. 55. Hackney AC, Aggon E. Chronic low testosterone levels in endurance trained men: the exercisehypogonadal male condition. J Biochem Physiol. 2018;1:103. 56. Hackney AC, Moore AW, Brownlee KK. Testosterone and endurance exercise: development of the “exercise-­hypogonadal male condition”. Acta Physiol Hung. 2005;92:121–37. 57. Arce JC, DeSouza MJ. Exercise and male factor infertility. Sports Med. 1993;15:146–69. 58. Bennell KL, Brukner PD, Malcolm SA. Effect of altered reproductive function and lowered testosterone levels on bone density in male endurance athletes. Br J Sports Med. 1996;30:205–8. 59. Hooper DR, Tenforde AS, Hackney AC. Treating exercise-associated low testosterone and its related symptoms. Phys Sportsmed. 2018:1–8. [Epub ahead of print]. 60. Hackney AC, Sinning WE, Bruot BC. Hypothalamic-­ pituitary-­ testicular axis function in endurance-­ trained males. Int J Sports Med. 1990;11:298–303. 61. Hooper DR, Kraemer WJ, Stearns RL, et al. Evidence of the exercise hypogonadal male condition at the 2011 Kona Ironman World Championships. Int J Sports Physiol Perform. 2018:1–22. [Epub ahead of print]. 62. Tenforde AS, Barrack MT, Nattiv A, et al. Parallels with the female athlete triad in male athletes. Sports Med. 2016;46:171–82. 63. Lane AR, Hackney AC. Reproductive dysfunction from the stress of exercise training is not gender specific: the “exercise-hypogonadal male condition”. J Endocrinol Diabetes. 2014;1:4. 64. MacConnie S, Barkan A, Lampman RM, et al. Decreased hypothalamic gonadotropin-releasing hormone secretion in male marathon runners. N Engl J Med. 1986;315:411–7. 120 65. McColl EM, Wheeler GD, Gomes P, et al. The effects of acute exercise on pulsatile LH release in high-mileage male runners. Clin Endocrinol. 1989;31:617–21. 66. Di Luigi L, Guidetti L, Baldari C, Fabbri A, Moretti C, Romanelli F. Physical stress and qualitative gonadotropin secretion: LH biological activity at rest and after exercise in trained and untrained men. Int J Sports Med. 2002;23:307–12. 67. Kujala UM, Alen M, Huhtaniemi IT. Gonadotrophin-­ releasing hormone and human chorionic gonadotrophin tests reveal that both hypothalamic and testicular endocrine functions are suppressed during acute prolonged physical exercise. Clin Endocrinol. 1990;33:219–25. 68. Safarinejad MR, Azma K, Kolahi AA. The effects of intensive, long-term treadmill running on reproductive hormones, hypothalamus–pituitary–testis axis, and semen quality: a randomized controlled study. J Endocrinol. 2009;200:259–71. 69. Hackney AC. The male reproductive system and endurance exercise. Med Sci Sports Exerc. 1996;28:180–9. 70. Hackney AC, Sharp RL, Runyan WS, Ness RJ. Relationship of resting prolactin and testosterone in males during intensive training. Br J Sports Med. 1989;23:194. 71. Blueher S, Mantzoros CS. Leptin in reproduction. Curr Opin Endocrinol Diabetes Obes. 2007;14:458–64. 72. Baylor LS, Hackney AC. Resting thyroid and leptin hormone changes in women following intense, prolonged exercise training. Eur J Appl Physiol. 2003;88:480–4. 73. Jürimäe J, Cicchella A, Jürimäe T, et al. Regular physical activity influences plasma ghrelin concentration in adolscent girls. Med Sci Sports Exerc. 2007;39:1736–41. 74. Hu Y, Asano K, Kim S, et al. Relationship between serum testosterone and activities of testicular enzymes after continuous and intermittent training in male rats. Int J Sports Med. 2004;25:99–102. 75. Deuster PA, Chrousos GP, Luger A, et al. Hormonal and metabolic responses of untrained, moderately trained, and highly trained men to three exercise intensities. Metabolism. 1989;38:141–8. 76. Le Panse B, Vibarel-Rebot N, Parage G, et al. Cortisol, DHEA, and testosterone concentrations in saliva in response to an international powerlifting competition. Stress. 2010;13:528–32. 77. Snegovskaya V, Viru A. Elevation of cortisol and growth hormone levels in the course of further improvement of performance capacity in trained rowers. Int J Sports Med. 1993;14:202–6. 78. Snegovskaya V, Viru A. Steroid and pituitary hormone responses to rowing: relative significance of exercise intensity and duration and performance level. Eur J Appl Physiol Occup Physiol. 1993;64:59–65. F. Lanfranco and M. A. Minetto 79. Passelergue P, Robert A, Lac G. Salivary cortisol and testosterone variations during an official and a simulated weightlifting competition. Int J Sports Med. 1995;16:298–303. 80. Minetto MA, Lanfranco F, Baldi M, et al. Corticotroph axis sensitivity after exercise: comparison between elite athletes and sedentary subjects. J Endocrinol Investig. 2007;30:215–23. 81. Sakakura N, Takebe K, Nakagawa S. Inhibition of luteinizing hormone secretion induced by synthetic LRH by long-term treatment with glucocorticoids in human subjects. J Clin Endocrinol Metab. 1975;40:774–9. 82. Chrousos GP, Torpy DJ, Gold PW. Interactions between the hypothalamic–pituitary–adrenal axis and the female reproductive system: clinical implications. Ann Intern Med. 1998;129:229–40. 83. Inder WJ, Jang C, Obeyesekere VR, Alford FP. Dexamethasone administration inhibits skeletal muscle expression of the androgen receptor and IGF-1--implications for steroid-induced myopathy. Clin Endocrinol. 2010;73:126–332. 84. Dufau ML, Tinajero JC, Fabbri A. Corticotropin-­ releasing factor: an antireproductive hormone of the testis. FASEB J. 1993;7:299–307. 85. Osterberg K, Karlson B, Hansen AM. Cognitive performance in patients with burnout, in relation to diurnal salivary cortisol. Stress. 2009;12:70–81. 86. Schulz P, Walker JP, Peyrin L, Soulier V, Curtin F, Steimer T. Lower sex hormones in men during anticipatory stress. Neuroreport. 1996;25:3101–4. 87. Francis KT. The relationship between high and low trait psychological stress, serum testosterone, and serum cortisol. Experientia. 1981;37:1296–7. 88. Nilsson PM, Moller L, Solstad K. Adverse effects of psychosocial stress on gonadal function and insulin levels in middle-aged males. J Intern Med. 1995;237:479–86. 89. Wheeler GD, Singh M, Pierce WD, et al. Endurance training decreases serum testosterone levels in men without change in luteinizing hormone pulsation release. J Clin Endocrinol Metab. 1991;72:422–5. 90. Minetto MA, Lanfranco F, Tibaudi A, Baldi M, Termine A, Ghigo E. Changes in awakening cortisol response and midnight salivary cortisol are sensitive markers of strenuous training-induced fatigue. J Endocrinol Investig. 2008;31:16–24. 91. Lucía A, Chicharro JL, Pérez M, Serratosa L, Bandrés F, Legido JC. Reproductive function in male endurance athletes: sperm analysis and hormonal profile. J Appl Physiol. 1996;81:2627–36. 92. Vaamonde D, Da Silva-Grigoletto ME, Fernandez JM, Algar-Santacruz C, García-Manso JM. Findings on sperm alterations and DNA fragmentation, nutritional, hormonal and antioxidant status in an elite triathlete. Case report. Rev Andal Med Deport. 2014;7:143–8. 93. Vaamonde D, Garcia-Manso JM, Hackney AC. Impact of physical activity and exercise on male 7 The Male Reproductive System, Exercise, and Training: Endocrine Adaptations 94. 95. 96. 97. 98. 99. 100. 101. 102. reproductive potential: a new assessment questionnaire. Rev Andal Med Deport. 2017;10:79–93. Wise LA, Cramer DW, Hornstein MD, Ashby RK, Missmer SA. Physical activity and semen quality among men attending an infertility clinic. Fertil Steril. 2011;95:1025–30. Gebreegziabher Y, Marcos E, McKinon W, Rogers G. Sperm characteristics of endurance trained cyclists. Int J Sports Med. 2004;25:247–51. Leibovitch I, Mor Y. The vicious cycling: bicycling related urogenital disorders. Eur Urol. 2005;47:277–86. Mastaloudis A, Leonard SW, Traber MG. Oxidative stress in athletes during extreme endurance exercise. Free Radic Biol Med. 2001;31:911–22. Astrand PO, Rodahl K. Circulation. In: van Dalen DB, editor. Textbook of work physiology: physiological basis of exercise, vol. 1986. New York: McGraw Hill Book Company; 1986. p. 170–5. Alessio HM. Exercise-induced oxidative stress. Med Sci Sports Exerc. 1993;25:218–224. Irvine DS. Glutathione as a treatment for male infertility. Rev Reprod. 1996;1:6–12. Vaamonde D, Diaz A, Rodriguez I. Preliminary results of trans-resveratrol as an effective protector against exercise-induced morphology abnormalities on mice sperm. Fertil Steril. 2011;96:S166–7. Vaamonde D, Da Silva-Grigoletto ME, Garcia-­ Manso JM, Vaamonde-Lemos R. Differences in sperm DNA fragmentation between high- and low-­ cycling volume triathletes: preliminary results. Fertil Steril. 2012;98:S85. 121 103. Child RB, Wilkinson DM, Fallowfield JL, Donnelly AE. Elevated serum antioxidant capacity and plasma malondialdehyde concentration in response to a simulated half marathon run. Med Sci Sports Exerc. 1998;30:1603–7. 104. Clarkson PM, Thompson HS. Antioxidants: what role do they play in physical activity and health? Am J Clin Nutr. 2000;72:637S–46S. 105. Hajizadeh Maleki B, Tartibian B, Chehrazi M. The effects of three different exercise modalities on markers of male reproduction in healthy subjects: a randomized controlled trial. Reproduction. 2017;153:157–74. 106. Hajizadeh Maleki B, Tartibian B. Resistance exercise modulates male factor infertility through anti-­ inflammatory and antioxidative mechanisms in infertile men: a RCT. Life Sci. 2018;203:150–60. 107. Hajizadeh Maleki B, Tartibian B. Combined aerobic and resistance exercise training for improving reproductive function in infertile men: a randomized controlled trial. Appl Physiol Nutr Metab. 2017;42:1293–306. 108. Hajizadeh Maleki B, Tartibian B. Moderate aerobic exercise training for improving reproductive function in infertile patients: a randomized controlled trial. Cytokine. 2017;92:55–67. 109. Hajizadeh Maleki B, Tartibian B. High-intensity exercise training for improving reproductive function in infertile patients: a randomized controlled trial. J Obstet Gynaecol Can. 2017;39:545–58. 8 Exercise and the Hypothalamus: Ovulatory Adaptations Angela Y. Liu, Moira A. Petit, and Jerilynn C. Prior Introduction As early as 1939, Hans Selye, who later received the Nobel prize for work on the endocrinology of the adaptation response, reported that muscular exercise was often a cause for “menstrual irregularities” in women [1]. Selye performed controlled animal experiments showing that whether or not exercise suppresses reproduction depends on the abruptness of exercise onset [1]. Forty years later, Shangold et al. [2] published the first prospective observational study documenting gradual shortening of the luteal-phase length with increased running activity in one woman with regular menstrual cycles. Despite these early observations indicating that subtle alterations of ovulatory function occur within cycles of normal length, the exercise science literature has since focused on the absence (amenorrhea) or presence (eumenorrhea) of menstrual flow in women athletes. The purpose of this chapter is to review the A. Y. Liu University of British Columbia, Medicine, Division of Endocrinology, Vancouver, BC, Canada M. A. Petit Activ8, LLC, St. Paul, MN, USA J. C. Prior (*) University of British Columbia, Medicine, Division of Endocrinology and Metabolism, Vancouver, BC, Canada e-mail: jerilynn.prior@ubc.ca subtle but clinically important ovulatory changes in response to exercise. Hundreds of cross-sectional studies report “athletic amenorrhea,“and inappropriately imply causal relationships between loss of flow and exercise. However, better-designed prospective studies observing normally ovulatory women and closely examining ovulatory function during progressively increasing exercise in reproductively mature women (subsequently termed “exercise training”) show only subclinical changes and no amenorrhea when exercise training is the only stressor [3–5]. Prevalent but subtle changes in ovulatory function are the first and most subtle hypothalamic adaptation to exercise training [2, 6]. Failure of hypothalamic adaptation in response to intense stressors such as starvation, psychological distress, illness, or rapidly increasing exercise results in significant disability. Overwhelming stress associated with excessive exercise training (see Chap. 9) and extreme nutritional imbalance (see Chap. 10) are discussed elsewhere in this volume. In this chapter, we describe the subtle alterations in ovulatory function that occur as a result of hypothalamic adaptation to exercise training and other “stressors.” We will also discuss the consequences of ovulatory disturbances, including infertility and a negative bone balance. Before beginning that discussion, however, it is necessary to define both the language and the physiological processes of ovulation. © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_8 123 A. Y. Liu et al. 124 The Ovulatory Cycle The words used to describe the release of an egg and the hormonal characteristics of a cycle in which that occurs need to be defined and described because both are usually obscured by the pervasive, yet imprecise notion that regular, normal-length cycles are always or usually normally ovulatory. We will start by defining the language of reproduction. Terminology In the exercise science literature, women are commonly inappropriately classified as “eumenorrheic” (which means “true menstruation!”) if their menstrual flow occurs monthly, or oligo/ amenorrheic if flow is sporadic or has been absent for 3 or more months [7]. However, cycles of normal length need to also be described by their postovulatory luteal phase or ovulatory characteristics. (Note that sometimes “anovulatory” is inaccurately used to mean oligo-amenorrhea). Ovulatory and cycle interval characteristics form a complex continuum (Fig. 8.1). This starts with the most normal cycle type, which is ovulatory with a normal luteal-phase length of 10–16 days (d) and a normal cycle length of 21–35 days [8]. This spectrum ends with the most disturbed ovarian function, which is amenorrhea, defined as the absence of flow for 3 or more months. Between these extremes, cycles that are normal in length may have a short (<10 d) or insufficient (normal length and estradiol levels, but low progesterone) luteal phases, or be anovulatory (normal estradiol but low progesterone levels). This latter is now termed “subclinical ovulatory disturbances.” Anovulatory cycles are ones in which cycle intervals may be short, normal, or long in length, but no egg is released and progesterone levels never meet or exceed 9.54 nmol/L (3 ng/mL) [9]. Fig. 8.1 A spectrum of cycle types starting at the top (I) with the most normal, which is of normal length and ovulatory with a normal luteal-phase length. The next cycle (II) is also of normal length and ovulatory, but has an insufficient but normal-length luteal phase. The third cycle (III), also of normal length, illustrates a short luteal-phase cycle. The fourth cycle (IV) is an anovulatory cycle of normal length, and the final cycle (V) is an anovulatory cycle that is longer than normal in cycle length (oligomenorrhea) I II III IV V 0 5 10 15 20 25 Days Flow Luteal phase 30 120 8 Exercise and the Hypothalamus: Ovulatory Adaptations Definitions Given the importance of clarity in science, it is useful to define the terms meant to describe cycle types when ovulatory status is not known. Eumenorrhea implies menstrual cycles that are normal in length, with flow occurring every 21–35 d [10]. When a woman’s flow occurs between 36 and <90 d, the term oligomenorrhea is appropriate. For cycle lengths of ≥90 d, women are classified as experiencing amenorrhea. Cycles of varying and abnormal lengths are called “cycle disturbances”; cycles of normal length but short absent or low progesterone levels are called “subclinical ovulatory disturbances.” Cycles are defined as having short luteal-­ phase lengths if ovulation occurs, but the time from ovulation to the day before the start of flow (luteal phase) is<10 d by quantitative basal temperature analysis [8, 11] or <12 d using the midcycle urinary luteinizing hormone (LH) peak as the indicator of luteal-phase onset. An inadequate or insufficient luteal phase means ovulation occurs and luteal-phase length is normal, but peak progesterone levels in the luteal phase are lower than usual (ideally ~45 nmol/L, [14.3 ng/ml]). If ovulation and subsequent corpus luteal formation do not occur, the cycle is anovulatory. Therefore, anovulation refers to cycles in which no eggs are formed (and released). “Subclinical ovulatory disturbances” are cycles that are normal in length, but have either short or inadequate luteal phases or are anovulatory. Cycles that are irregular or abnormal in length and about which ovulatory characteristics are (usually) unknown should be termed “cycle disturbances” and include polymenorrhea (cycles shorter than 21 d) as well as oligomenorrhea and amenorrhea. I mprecise Language About Reproduction Two terms are sometimes used that have problems of precision: “eumenorrheic/eumenorrhea” and “anovulatory/anovulation.” There are several problems with classifying women as eumenorrheic. The term has been applied to women who “experienced at least 10 menstrual periods per year” [12], even though this would give an aver- 125 age cycle length of 36.5 d (which is abnormally long) and that is oligomenorrhea. Another difficulty with the term, eumenorrheic or eumenorrhea, is that it presumes that all cycles of normal length display the same ovulatory and hormonal characteristics. Data from our 1-year prospective study in ovulatory women of varying exercise habits [4] showed that normal-length cycles could as easily be anovulatory as ovulatory. In that study, all of the anovulatory cycles were normal in length. Therefore, a further erroneous assumption often made in the literature is that only long or short cycles are hormonally abnormal. The term “anovulatory/anovulation” menstrual cycle is also often misused. Researchers often assume a woman is “anovulatory” if she reports that her cycles are long or irregular—that may or may not be the case. Likewise, the term “anovulation” is commonly used as a synonym for amenorrhea or oligomenorrhea because women whose periods are long or have stopped (unless they have become pregnant) are usually not ovulating. In short, classifying women only by their cycle intervals implies that the reproductive system works in an on-off or mechanical manner, rather than displaying the broad spectrum of potential responses described above. Classification of women’s cycles needs to include the entire range of cycle and ovulatory types, because a distinctly different hormonal profile is present in each case. In addition, the variability and hormonal physiology of cycles, even those of normal length, are important to understand. Physiology Just as cycles vary in interval and ovulatory characteristics, so does the cascade of signals from the hypothalamic gonadotrophin-releasing hormone (GnRH) nucleus to the pituitary gonadotrophin-­ producing cells. Pituitary messages to the ovarian follicle also change, as do hormones from the ovary that give feedback to the pituitary and the hypothalamus. What follows is an effort to clarify the cycle manifestations of the hypothalamic changes described in earlier chapters (i.e., Chaps. 1 and 4). 126 varian Hormone Levels During O the Normal Cycle An ovulatory menstrual cycle is characterized by systematic and major changes in the levels of estradiol prior to ovulation (follicular phase) and variations of both estradiol and progesterone post-ovulation (luteal phase). Follicular-phase estradiol levels during and just after flow average 60–200 pmol/L (levels that are similar to those in children and men). Estradiol levels subsequently rise over the next 7–18 d to a peak just prior to ovulation that is, on average, 220–250% above the follicular-phase baseline [13]. There is then a decrease to about 100% above baseline for most of the luteal phase before estradiol levels again decrease to low levels just prior to menstruation [13]. In contrast, progesterone levels, which remain low during the entire follicular phase (~0.5–2.0 nmol/L, similar to levels in children and men), increase after ovulation to over 1400% of follicular-phase baseline values. Progesterone levels produced by one corpus luteum remain elevated over 1000% above baseline during the 10–16 d of the luteal phase [13]. The production of estradiol and progesterone is coordinated by, and ultimately dependent on, the timing and magnitude of GnRH pulsatility in the hypothalamus. GnRH stimulates the gonadotrophins, LH, and follicle-stimulating hormone (FSH), to be released from the pituitary. LH peaks at midcycle, and directly triggers follicle rupture and egg release. FSH plays an important role in recruiting intermediate-sized follicles and stimulating the dominant follicle that eventually ovulates. In addition, FSH increases LH receptors on ovarian granulosa cells. GnRH, LH, and FSH are all in feedback regulation by estradiol and progesterone levels. Also, FSH production is actively suppressed by inhibin, a polypeptide hormone that is key in perimenopause [14] but whose potentially important role in reproductive physiology remains poorly understood [15]. ormonal Profile Changes During H Disturbed Cycles Hormonal characteristics of cycles related to their length will be briefly discussed followed by the hormonal characteristics of cycles that have A. Y. Liu et al. disturbed ovulatory characteristics. Although few studies have systematically measured estradiol levels in cycles that are short or long, the generalization that shorter cycles have higher estradiol levels is supported by a study in which hormone levels were measured daily during 68 cycles [16]. That study documented that shorter follicular-­ phase lengths were associated with statistically higher follicular and whole cycle estradiol levels [16]. The logic of this observation is that the more estradiol stimulation of the endometrium, the more likely it is to shed causing bleeding. The opposite is true of long cycles—less estradiol stimulation of the endometrium leads to delayed shedding and flow. The hormonal characteristics of cycles with disturbed ovulation are less clear. The common feature of all disturbed cycles is the lower amount and/or duration of progesterone production. Estrogen and androgen productions are highly variable in individuals with ovulatory disturbances. Evidence for high estrogen levels with anovulatory cycles is most clearly found in studies of women shortly after puberty [17] and in perimenopausal women [18]. In both instances, estrogen levels exceed the midcycle peak equivalent levels for prolonged periods of time. Androgen excess, which is associated with anovulation, is also associated with high estradiol levels [19], obesity, insulin resistance, and varying degrees of hirsutism. Evidence that estradiol levels may be normal in anovulatory cycles comes from our observational prospective study [4]. In that group of initially ovulatory women (in whom perimenopause and androgen excess were excluded), the cycles without ovulation were normal in length, and the women who had entirely normal ovulation did not differ in mean estradiol level (measured twice in two cycles a year apart) from the women who experienced anovulation. This flies in the face of the expectation that cycles with disturbed ovulation will have low estradiol levels as had been observed in four women studied by Sherman and Korenman [20]. Sowers et al. also have reported a few days of lower mid-follicular estradiol levels in premenopausal women with disturbed ovulation [21]. However, several other authors in addi- 8 Exercise and the Hypothalamus: Ovulatory Adaptations tion to ourselves have not observed consistently low estradiol levels associated with anovulation [22, 23]. By contrast, the estradiol levels were minimally, although significantly lower in cycles without progesterone levels above 9.54 nmol/L in a single normal-length cycle study in over 3000 women in Norway [9]. In summary, disturbances of cycle interval are often associated with abnormally low or high estradiol levels (inversely related to cycle length), but cycles with ovulatory disturbances may have high, normal, or low estradiol levels and rates of production. Documentation of Ovulatory Function This section describes the currently available methods for documenting ovulatory function and the advantages and disadvantages of each. Our primary focus will be to describe the use of “Quantitative Basal Temperature” (QBT), which we have found to be the best available method for continuous, longitudinal monitoring of ovulatory function. urrently Used Indirect Ovulation C Detection Methods All of the currently available methods for assessing ovulation are indirect, except actual visualization of extrusion of a secondary oocyte from the ovary. The closest to an indirect “gold standard” for ovulation is serial ovarian ultrasounds observing a dominant-sized follicle that “disappears” because it has ruptured and released an egg. Because ovulation requires an LH surge and progesterone levels do not rise if ovulation does not occur, serum or urinary measures of the midcycle LH peak and/or progesterone levels are often used as indicators of ovulation. One method is to perform serial samples of serum or urine daily during the midcycle to detect the LH peak. Alternatively, in the week prior to menses, serum (or plasma) samples showing levels of progesterone of ≥9.54 nmol/L (≥3ng/mL) are indicative of ovulation. The postovulatory increase in progesterone can also be measured in spots of whole blood [24], urine, or saliva [25] or by its effect to 127 increase core temperature or to inhibit the stretch/ elastic characteristics of estrogen-stimulated cervical mucus (although this latter effect has not been scientifically evaluated to date). An estradiol peak is necessary to trigger the midcycle LH peak. Therefore, another indirect assessment of ovulation involves collecting estradiol levels daily with serum samples. Samples must be taken until an estradiol level doubles the preceding level and over 750 pmol/L (by usual assays) is documented. However, midcycle peak estradiol levels may occur and not be followed by an LH peak or by ovulation in premenopausal (as in perimenopausal) women [23, 26, 27]. Therefore, an estradiol peak level is not a specific test of ovulation, nor is the stretch of cervical mucus that estradiol stimulates. To a lesser degree, the same lack of specificity is also true of an LH peak [27]. imitations of Available Methods L for Diagnosis of Ovulation Disturbances Serial sampling of blood, saliva, or urine is required to adequately document all of the important ovulatory characteristics (including whether ovulation occurred, as well as luteal-phase adequacy and length) of a single cycle. Using these methods to document several consecutive cycles is very labor-intensive, invasive, expensive, and imposes a high degree of burden on participants. Continuous longitudinal documentation of hormone levels in large numbers of women is, therefore, virtually impossible to obtain using these sampling techniques [3, 5, 27]. Similarly, although formerly endometrial biopsy analysis of histological change related to progesterone was considered definitive for luteal-phase adequacy and length, it has a ±2-d SD and is not useful [28]. Finally, serial ultrasound assessments (to show a growing follicular cyst that enlarges to over 18mm and then disappears) are now considered the indirect “gold standard” indicator of the occurrence of ovulation [25], but they lack convenience and reasonable cost for most studies. The logical question is: why not measure ovulatory characteristics during one cycle and then just monitor cycle intervals over the necessary 128 period of time? Could you not infer that the subsequent cycles, if they are regular and normal in length, are similar in ovulatory status? That would be an accurate strategy if women’s cycles were as stable in ovulatory characteristics as they are in cycle interval. However, ovulatory characteristics are highly variable over time within women [4, 8, 29, 30]. For example, Hinney et al. [29] documented “corpus luteum insufficiency” by a late luteal-phase progesterone level below 25 nmol/L in 109 women of whom only 55, when tested in the following cycle, continued to show corpus luteum insufficiency. Likewise, 5 years after our intensive monitoring of continuous cycles for 1 year in 66 women, cycle lengths (in ≥3 cycles) correlated well with previous ones (r = 0.68, P < 0.05). However, luteal-phase lengths correlated considerably less well (r = 0.39, P = <0.05) [30]. Furthermore, as this chapter will subsequently document, ovulatory disturbances caused by hypothalamic adaptation occur rapidly and as quickly revert to normal ovulation. Thus, studies that measure ovulatory characteristics in only a few cycles or monitor cycles discontinuously (such as every other or every fourth cycle) are not likely to detect ovulatory disturbances (in general) and particularly not likely to document those related to hypothalamic adaptation to exercise. That is especially true if cycle characteristics are documented only in the cycle before exercise intensity is again increased, as has been done in two prospective studies [3, 5]. At least 6 months of continuous sampling, in which both ovulation and luteal-phase lengths are assessed, are necessary to adequately characterize a sedentary, weight-stable woman’s menstrual and ovulatory characteristics [31]. In exercising women, it is even more critical to provide a robust baseline from which to examine potential changes associated with exercise training. For all of these reasons, a noninvasive, inexpensive, and “habitforming” method for documenting ovulatory characteristics is necessary. uantitative Basal Temperature (QBT) Q Daily basal (meaning first thing after wakening in the morning, fasting, and when metabolism is A. Y. Liu et al. stable) oral temperatures (often referred to as BBT) potentially allow continuous, longitudinal research into ovulatory characteristics to be conducted. High levels of progesterone during the luteal phase increase the basal temperature. This increase begins to be significant approximately 24–48 h after the LH peak [11]. A monophasic set of basal temperatures during one cycle, in which our least-squares program (Maximina®) detected no significant shift, characterized an anovulatory cycle with progesterone levels that did not rise sufficiently to alter temperatures. A biphasic cycle is indicative of ovulation [8], and the day of the significant temperature shift can be used to define the onset of the luteal phase [11]. In ovulatory cycles, the increased progesterone levels raise the basal temperature during the luteal phase by approximately 0.2–0.3 °C. However, BBT was a clinical tool before it was a research method. Therefore, the early studies utilizing BBT as a method of detecting ovulation had a number of problems, including that women might take their temperature at different times of day, women had difficulties reading or shaking down the older mercury thermometers, women were expected to plot their own temperatures as a graph (which often resulted in inaccuracies of graphing), and the temperature patterns were evaluated for the presence or absence of ovulation using non-quantitative methods and often “eyeball” or equally nonreproducible methods [32]. Finally, even when more systematic methods of assessing changes in temperatures were described [33], insufficient data relating the temperature shift to hormonal data were available. In our laboratory, these problems have been resolved by better instruction of women about what the factors in addition to fever that alter the basal temperature (such as awakening earlier or later than usual, or being up in the night) and providing a form on which to record these factors. In addition, we asked women to take their temperature with a digital thermometer reading to two decimal places and to record temperatures in a list, rather than plotting them on a graph. We then devised and applied a 8 Exercise and the Hypothalamus: Ovulatory Adaptations computer program (Maximina®) of leastsquare analysis to each cycle of temperature data and showed it to be valid against the independently assessed serum LH peak (r = 0.88) [11]. At the same time, we validated the “mean temperature” method of Vollman [8, 11]. This more scientific method we named “Quantitative Basal Temperature”, so we could differentiate it from the crude and unscientific BBT methods used in the past. Thus, we believe we have transformed the previously inaccurate and unreliable BBT method into a scientific tool for documentation of ovulatory characteristics. Furthermore, it is a method that can be easily taught, requires only a relatively inexpensive and durable digital thermometer, and is one that interested women can and will consistently use [4, 34] for lengths of time exceeding a year. Taking of basal oral temperature quickly becomes a habit. However, for this to happen, it does require the interest and commitment of women and of those teaching them. The major difficulty with widespread use of the QBT method is its lack of accuracy in those whose time of waking and sleeping is variable (e.g., those on shift work, with small children, or students—although it was robust to time of awakening in one study [35]). A simpler method, not dependent on a stable life pattern, and requiring less commitment from women, is needed for documentation of ovulatory characteristics in longitudinal studies and for epidemiology. Hypothalamic Adaptation and Ovulatory Function The neuroendocrine physiology of adaptation to exercise and other stressors is complex and not yet completely understood. A review article published more than 10 years ago continues to reflect our current understanding on the subject [36]. It has also been covered by earlier chapters in this volume, and therefore will be reviewed only briefly here. The hypothalamus functions to maintain internal homeostasis in response to internal and external factors. 129 Numerous influences, such as ambient and core temperature changes, energy balance changes, illness (which alters eating and sleeping patterns and may cause elevated temperature levels), and psychological stress, can directly or indirectly alter the pulsatile secretion of GnRH and thus change subsequent reproductive function [36]. The premise of this chapter is that the first and most subtle adaptive responses to exercise training occur during the premenstrual phase of the cycle with a range of ovulatory disturbances (Fig. 8.1) that all result in decreased total exposure to progesterone. As discussed, studies that examine hypothalamic control and the subtle changes that lead to shortening of luteal-phase length with exercise training require long-term, continuous monitoring of ovulatory function. Ovulatory disturbances in response to exercise training can be viewed as an adaptation to the increased physiological and perhaps psychological stresses of the exercise and are not part of a disease process. The adaptation model suggests these four principles: 1. Ovulatory disturbances are caused by a hypothalamic process that is conservative, e.g., protective of or saving energy for the individual. 2. They are induced by a variety of physical and psychological “stressors,” which act through a common mechanism and manifest similar changes. 3. There are gradients of change in response to the severity or intensity of the “stress” or “threat.” 4. The adaptive changes reverse to the normal baseline steady state when the “threat” is lessened or eliminated, or the individual has sufficient time and is able to adapt. Evidence for these points will be described in the following sections. The specific ovulatory adaptations to exercise training, including the gradients of change and reversibility, will be described in the section “Adaptations to Exercise Training” of this chapter. A. Y. Liu et al. 130 Hypothalamic Adaptive Processes Evidence that the subtle alterations that lead to shortening of luteal-phase length are controlled by the hypothalamus is largely circumstantial, because altering hypothalamic function biochemically or with direct nerve cell stimulation is impossible in humans. The strongest evidence that the hypothalamus controls changes in ovulatory function comes from the similar pattern of responses during exposures to a whole range of psychological and physiological stressors. Corticotrophin-releasing hormone (CRH) discharge increases when any internal or external environmental signal is perceived as stressful (as shown schematically in Fig. 8.2). The increased CRH may either directly or indirectly (via the β-endorphin system) slow the hypothalamic pulsatile release of GnRH [38] and, therefore, decrease pulsatile LH release. Fig. 8.2 Processes through which physical (including exercise and illness), emotional, or nutritional challenges cause increased release of CRH from the hypothalamus. These factors suppress the reproductive system and stimulate the adrenal axis. ACTH corticotrophin; LH luteinizing hormone. (Modified from Prior [37]) Because the pulses of LH stimulate estradiol and androgen secretion, they provide an essential precursor to ovulation. A systematic review by Hakimi and Cameron [39] on the effect of exercise on ovulation proposed mechanisms by which vigorous exercise disrupted ovulation in women with normal and low body mass index, and by which exercise restores ovulation in overweight and obese women. They describe a U-shaped association of exercise with ovulation: increases in ovulatory disturbances with increased exercise especially in women or normal or lower weight but decreases in (pre-existing) ovulatory disturbances with exercise in overweight women. This is based on ten interventional and four observational cohort studies, which showed that greater than 60 min per day of heavy exercise was associated with an increased risk of anovulation, while vigorous exercise of 30–60 min per day was associated Threats/stresses: • Physical • Emotional • Nutritional • Overtraining Corticotrophin-releasing hormone(CRH) ? ↑ β-endorphin (neurotransmitters) LH pulse frequency ↑ ACTH Subclinical ovulatory disturbances • Anovulation • Short luteal phase ↑ Cortisol ↓ GnRH Cycle disturbances • Amenorrhea • Oligomenorrhea Normal estradiol ↓ Progesterone Accelerated bone loss ↓ Estradiol and ↓ Progesterone 8 Exercise and the Hypothalamus: Ovulatory Adaptations with a reduced risk of anovulation. Seven studies examining exercise in overweight and obese women with polycystic ovarian syndrome found that exercise was associated with improved ovulatory function. Notably, one limitation in these studies is the lack of assessment of rate of increase in exercise intensity. Many of the cross-­ sectional studies simply observe current exercise duration and intensity. Evidence documents that the rate of progression of exercise training is an important modulator as well [1, 40]. Reproductive and primarily ovulatory changes are “conservative” for the individual, because through multiple pathways, they effectively prevent pregnancy when the woman is unable to physically or emotionally support a healthy process. They are also conservative of energy because less progesterone production, which decreases the otherwise increased core temperature, means women can consume about 300 fewer dietary calories and, like women with normal ovulation, still ensure energy balance [41]. 131 In humans, reversible, modulated suppression of reproduction during illness was documented by lower than normal LH levels in gravely ill, hospitalized postmenopausal women; LH levels recovered as they improved [45]. Similarly, a prospective study in Japanese nursing students showed regular and apparently ovulatory cycles with more frequent ovulatory disturbances during the stressful school year than in the summer break [46]. Weight loss is known to be one of the most powerful physiological hypothalamic stressors [47, 48]. An experimental protocol involving fasting for 3 d in the late follicular phase appears to be more disruptive of follicle development and more likely to suppress LH pulsatility in women who are initially very lean than in those who have normal body weights and fat [49]. Active women with amenorrhea, like over-­ trained athletic men [50], have increased basal levels of cortisol [51] and blunted cortisol responses to exercise [52, 53]. Berga et al. [52] reported high 24-h cortisol levels in those with Stress Mechanism hypothalamic forms of oligo-/amenorrhea comSelye [1] observed about 70 years ago that the pared with normally menstruating women. This adrenal glands were hypertrophied when various hypercortisolemia was not observed in women kinds of stressors interrupted estrus in rats. He with other reasons for disturbed cycles, such as also observed similar patterns of response of the hyperandrogenism or hyperprolactinemia. A few ovaries and the adrenals to excessive exercise, to women initially deemed to have hypothalamic interference with normal diet, and to emotional amenorrhea subsequently ovulated during the stressors. More recently a strong relationship was study and were shown to have concomitantly also documented between social stress and non-­ reduced levels of cortisol [52]. Ding et al. [51] ovulation in nonhuman primates. Subordinate could similarly predict women whose cycle interfemale monkeys experienced 16.5% of cycles as vals would subsequently become normal because non-ovulatory, whereas dominant females over their cortisol excretion was decreased. the same time period and in the same conditions High cortisol secretion or urinary excretion experienced only 3.5% of their cycles as anovula- has become a useful marker of hypothalamic tory [42].The subordinate monkeys at autopsy adaptive responses to stressors including exerhad very enlarged adrenal glands [42]. Cortisol cise, because all stressors apparently act through excess, which was similar to levels seen in the hypothalamic CRH pathway. Therefore, studwomen under stress, significantly increased the ies in both humans and nonhuman primates demmetabolic clearance of progesterone as well as onstrate increased cortisol levels simultaneously increasing LH pulse amplitude in experimental with decreased LH pulsatility and/or disturbed studies by Kowalski et al. [43]. This research ovulatory function during reproductive showed that monkeys who were exposed to ­disturbances coinciding with a variety of stressinduced hypercortisolism had lower luteal-phase ful situations. serum progesterone levels and more ovulatory It should be noted that, although hypothalamic disturbances [43, 44]. disturbances of ovulation characterized by lower A. Y. Liu et al. 132 pulsatile release of LH are probably the most common cause for the menstrual cycle disturbances reported in athletes, short luteal-phase cycles or an ovulation associated with androgen excess (and with high, rather than low, LH levels) [54, 55] can also be documented. High androgen and LH levels have been described in swimmers with amenorrhea [54]. In addition, defects of the large corpus luteum cells have been postulated to cause lower luteal-phase progesterone levels, although LH pulsatility and estradiol levels are both normal [29]. Energy Conservation Reversible cycle disturbances are termed “functional” (that implies psychological) and are not a disease. When discussing ovulatory disturbances as protective against excess energy expenditure, the severity of the disturbance is proportional to the amount of energy conserved. Amenorrhea in women without an extreme eating disorder may be relatively less threatening than anorexia, because compared with menstruating women, it appears to lower BMR only 17% [56]. Anovulatory cycles, which are normal in length, are also less metabolically costly to maintain than ovulatory cycles and prevent the risk of pregnancy with its high-energy demands. The basal temperature increase during the luteal phase raises metabolic rate. Barr et al. [41] documented that women’s dietary intake was increased approximately 300 kcal/d during the hormonally confirmed luteal phase of cycles compared with anovulatory cycles in the same group of women; all were without exercise or weight changes during the six-cycle study [41]. A shortened luteal-phase length (in contrast to anovulation) occurs in response to the least threatening intensity or kind of stressor. Energy demands are higher when the luteal-phase length is shortened than they are in anovulatory cycles, due to up to 9 d of progesterone-related temperature elevation in the former. We believe that shortening of luteal-phase lengths is the most common adaptive response to stressors, such as weight loss, emotional stress, illness, or exercise training. It is of importance that, despite the minimal alteration of ovarian physiology, fertilization and implantation of the egg are still prevented by subclinical short luteal phase and luteal insufficient cycles. ynergism or Interactions Among S Factors Influencing Ovulatory Function The concept of adaptation with a common hypothalamic change caused by many different stressors implies that the response to one, such as exercise, would depend on the current state of other factors, such as energy balance or emotional stress. Therefore, it is important to consider those factors that are known to influence ovulatory function and to acknowledge that individuals may respond differently to any given stress depending on the presence of many personal variables. The adaptive response is altered by such factors as the individual’s current energy balance, underlying characteristics of the individual (i.e., levels of reproductive maturation, weight, and emotional well-being), intensity of the threat, and the rapidity with which it is introduced. Multiple emotional and psychological stressors, weight loss or restrictive eating, and the need to feel “in control” all are often perceived as stressful by the hypothalamus and influence reproductive function (Fig. 8.3). These stressors all appear to act through the common hypothalamic CRH pathway. Energy Balance It is likely that exercise and other stressors affect LH pulsatility through their influence on energy balance [57]. Other chapters in this volume discuss this (see Chaps. 11 and 17). We postulated in 1982 that hypothalamic insulin receptors might provide a common signal [6]. Those who are ill or over-exercising would have decreases in their insulin levels as a consequence of negative caloric balance. It is well accepted that severe weight loss or an extreme energy deficit, such as with anorexia nervosa, suppresses reproductive function. In such extreme cases, CRH 8 Exercise and the Hypothalamus: Ovulatory Adaptations 133 Emotional stress/threats Need to feel “in control” Hypothalamic-gonadal suppression Compulsive exercise Stress fractures Increased musculoskeletal injury Binge/purge syndrome Cognitive dietary restraint ↓ Estradiol and ↓ Progesterone Weight loss Infertility ↓ Libido Ovulatory disturbances Fig. 8.3 Interrelationships among multiple factors (stress, compulsive exercise (associated with an increased risk for relative energy deficiency), and cognitive dietary restraint) that appear to be causally related to the development of ovulatory disturbances levels are high [58] and amenorrhea will likely result. More subtle reproductive disturbances often occur when the relative threat is less intense, but the conditions that facilitate pregnancy are still not optimal. For example, ovulatory disturbances may occur with healthy weight loss or dieting [59], as well as when recreational exercise or emotional stress increase. In each case, the greater the need for energy conservation, the more severe the ovulatory or cycle length disturbance [48, 60]. In 2014, Mountjoy et al. as part of an International Olympic Committee Expert Group presented the concept of relative energy deficiency in sport (RED-S) (Fig. 8.4). This is a more physiological and comprehensive alternative to what was called the “female athlete triad” that refereed only to athletic women [61]. RED-S is a syndrome characterized by impaired physiological function in areas such as metabolic rate, reproductive function (that, for women means menstrual cycle and ovulatory changes), bone health, immunity, protein synthesis, and cardiovascular health. It is due to an imbalance between dietary energy intakes relative to energy expenditures; when expenditure exceeds intake, this results in a relative energy deficiency. The syndrome of RED-S is applicable to both women and men and takes a physiological rather than a disease approach to the multitude of changes related to exercise training. The magnitude of energy deficit affected the frequency of menstrual cycle and ovulatory disturbances in healthy women [63]. The average percentage of energy deficit, in a study of untrained, regularly cycling and ovulating women aged 18–30 y followed over four menstrual cycles, was the major predictor of the frequency of menstrual cycle/ovulatory disturbances even when adjusted for weight loss. Luteal-phase disturbances, although they only vaguely defined their documentation, were the most frequently observed reproductive changes. However, there were no differences by the degree of relative energy insufficiency in the development of anovulatory cycles or oligomenorrhea [63]. When these authors defined energy availability as energy intake minus A. Y. Liu et al. 134 Immunological Menstrual function Gastrointestinal Triad Cardiovascular Bone health RED-S Endocrine Psychological* Growth + development Metabolic Hematological Fig. 8.4 Potential health effects of relative energy deficiency in sport (RED-S). This is applicable to both men and women athletes. Note that ∗Psychological conse- quences may precede or result from RED-S. (Modified from [61] and the concept adapted from the original idea of N. Constantini [62]) exercise energy expenditure divided by kilograms of lean body mass and looked for associations with ovulatory disturbances in 91 exercising women, they found it discriminated clinical menstrual status (e.g., amenorrhea vs. regular menstrual cycles) but not subclinical ovulatory disturbances [64]. This, again, highlights the distinction between menstrual cycle length changes that are clinically obvious and the silent and more prevalent ovulatory disturbances. Cognitive Dietary Restraint Subtle ovulatory disturbances also occur with cognitive dietary restraint (also called “eating restraint”), a psychological attitude in which women feel they must limit food intake to avoid obesity. Women who are classified as highly restrained (based on the Three Factor Eating Questionnaire [65]) are very conscious of their food intake, but they do not necessarily consume 8 Exercise and the Hypothalamus: Ovulatory Adaptations 135 fewer calories than weight- and age-matched ponectin; and decreased triiodothyronine and kiscontrols who are not restrained [66, 67]. Because speptin [72]. Progesterone therapy in an RCT in maintaining or achieving their desired weight is menopausal women caused a small but signifiso important to their emotional well-being, eat- cant increase in the level of free T4 [73]. ing is associated with psychological stress for Collectively, these changes appear to suppress women with cognitive dietary restraint. A very the hypothalamic–pituitary–ovarian axis in an early study using the Eating Restraint Scale of adaptive, graded manner [72]. the Three Factor Eating Questionnaire showed It is probable that the effect of cognitive that women with higher scores were more likely dietary restraint on ovulatory function is medito have short luteal-phase cycles [68]. Three ated through hypothalamic adaptation pathways. studies from our laboratory also examined ovula- The evidence that subtle ovulatory disturbances tory function and eating restraint in normal are more common among those with greater cogweight, regularly cycling, and ovulatory women nitive dietary restraint, despite similar energy who varied in their usual activity levels [69] and intakes and expenditures, emphasizes that hypoin regularly cycling vegetarian and non-­ thalamic ovulatory disturbances may result from vegetarian women [70]. A more recent study in relatively minor psychological as well as physiyoung adult women (most of whom were post-­ ological stressors. secondary students) showed that those with higher eating restraint scores had more ovulatory disturbances and higher 24-hour urine-free corti- Hypothalamic Reproductive sol levels [71]. The frequency of subclinical ovu- “Maturation” latory disturbances and the degree of cognitive dietary restraint were associated with less posi- Another variable influencing the ability of the tive changes in bone mineral density, although hypothalamic/pituitary/ovarian system to respond cortisol did not appear to modulate that relation- to stressors is its relative maturity. For example, ship [71]. In all of these studies, the Restraint the majority of menstrual cycles are anovulatory Scale of the Three Factor Eating Questionnaire in the first year after menarche [8]. However, on [65] was administered initially, and menstrual average, women do not develop the highest rate cycle characteristics were documented prospec- of ovulatory cycles until they are approximately tively over three or six cycles or 2 years, respec- 12 years after menarche [8] (or gynecologic age tively. In all studies, women in the highest versus 12). This implies that some are still gynecologilowest tertile of restraint were significantly more cally immature. It fits with the adaptation hypothlikely to experience a short luteal phase or anovu- esis that those whose hypothalamic–reproductive latory cycle. These findings could not be attrib- axis has not yet become sturdily and regularly uted to differences in energy intakes, exercise ovulatory are more likely than those with mature levels, or body mass index (BMI is weight in kg reproductive patterns to respond to stress with divided by height in m2) levels. Women with eat- altered cycle lengths as well as with ovulatory ing restraint did not differ in BMI, weight, energy disturbances [46]. intake, activity levels, or cycle lengths from the One of the first studies documenting the reproless restrained women in each respective popula- ductive hormonal characteristics of young athtion [69–71]. Since cycle intervals were unal- letes showed that both swimmers and controls tered, none of these women would have known had short luteal-phase cycles, but in swimmers, their ovulation was disturbed. the luteal phase was even shorter than in ­sedentary Changes in metabolic hormone levels have controls [74]. Although participant numbers were been documented related to resting energy expen- small, these data confirm the more extensive data diture. Metabolic alterations include growth hor- of Vollman [8] that teenagers are susceptible to mone resistance and reduced IGF-1 concentrations; subtle disturbances of ovulation. Young runners increased cortisol, ghrelin, peptide YY, and adi- (gynecologic age <10 year, mean chronologic A. Y. Liu et al. 136 age 20 year) are also more likely to have disturbed folliculogenesis and decreased estradiol, progesterone, gonadotrophins, and testosterone levels than are gynecologically mature women (gynecologic age >15 year, mean chronological age 31 year) [75]. Therefore, data suggest that the combination of more intense training and an immature hypothalamus are potentially additives in suppressing reproduction in young women. Mature gynecological aged women who begin exercise or intensify training only experience ovulatory and not cycle-length changes. However, evidence suggests, although we do not yet have appropriate experiments to document it conclusively, that a woman in her 20s who is initially only intermittently ovulatory and begins to exercise or intensifies exercise training may well develop cycle as well as ovulatory disturbances. This young woman, with weight loss or emotional distress added to exercise training, would likely develop oligomenorrhea or amenorrhea. Evidence says that age at menarche is influenced by the energy imbalance related to intense exercise training [76–78]. Although genetic factors also have a strong influence on menarcheal age [79], dancers and gymnasts who experience lower energy availability are more likely to have delayed menarche compared with their sedentary sisters even though they are genetically very similar. Puberty involves maturation of axillary and pubic hair as well as breast enlargement and areolar/nipple maturation. Interestingly, when young athletes are forced (often because of injury) to interrupt their gymnastics or dance training, rapid development through one or more of the Tanner breast stages commonly occurs [78, 80]. Anorexia nervosa commonly occurs in women and during puberty. Weight loss and young age may make them more vulnerable to anorexia. In a similar manner, they will likely be more prone to exercise effects on ovulatory function, especially if exercise is combined with restricted energy intake or psychological performance pressure from coaches and parents. It is also probable that women experiencing reproductive and ovulatory disturbances in response to stress when younger will be more susceptible to exaggerated stress responses throughout life [81]. The pubertal maturation of the breast is primarily dependent on ovarian hormones, with little or no influence of adrenal steroids. By contrast, pubic hair maturation can proceed with the normal adolescent increases in adrenal androgen secretion, without significant increases in ovarian hormones. Discrepancy in the degree of Tanner stage breast compared with pubic hair maturation is probably a clue to hypothalamic adaptive changes related to exercise training and/or other stressors. Warren et al. [78] reported that pubic hair development occurred at a normal age in young women dancers, but there was a trend to delays in breast development and age at menarche. Clinical data from ovarian hormone treatment of male-to-female transgender individuals [82] and observations during a prospective study of puberty [83] both suggest that normal breast development to the fully mature Tanner stage V breast will not be reached without adequate exposure to ovulation and thus to high progesterone levels. tress Intensity and the Rate S of Increase in Stress Intensity Whether ovulation becomes disturbed partially depends on the intensity of the stress and partly on the rate of introduction of that stress. For example, in one study all rats responded to “inescapable” shock by suppressed gonadotrophin secretion [84], whereas only some rats were susceptible to the relatively less threatening stress of gradually increased endurance exercise [1]. Hans Selye coined the term “general adaptation syndrome” and published early controlled trials of exercise and energy restriction stress on rats [1]. Selye’s experiments showed a dramatically different response to gradually increasing exercise compared with rapid imposition of exercise training (or caloric restriction) (Fig. 8.5). Animals which started running at 3.5 km/d ­developed anestrus (the rat equivalent of amenorrhea) with interstitial atrophy, few mature follicles, and increased weight of their adrenal glands. A second group of rats gradually increased exercise intensity to reach 3.5 km/d over 4 weeks (Fig. 8.5). Even though 8 Exercise and the Hypothalamus: Ovulatory Adaptations Fig. 8.5 Illustration of the concept of the “general adaptation syndrome” developed by Hans Selye. Exercise was introduced abruptly or gradually in rats randomized to one or the other group. The photomicrographs show ovulatory adaptation by normal interstitum and follicular development in rats with gradual increase in exercise. Abrupt introduction of exercise led to anestrus (the rat equivalent of amenorrhea), interstitial atrophy, and development of only a few mature follicles. (Data redrawn from Ref. [1]) 137 Exercise Abrupt Gradual 30.5 cm wheel 20 r.p.m. Initial 15 min - 3 x day 0.86 km/day 60 min - 3 x day 3.5 km/day 2 weeks 30 min - 3 x day 1.7 km/day ” 4 weeks 60 min - 3 x day 3.5 km/day ” Normal estrus Anestrus OVARY at 3 mo. Normal interstitium the rats in the second group maintained the same level of exercise intensity as the first group for 2 of the 3 months, reproductive function remained normal, and ovarian follicle development was appropriate. Similar differences in response were observed in rats treated with rapid “semi-starvation” compared with gradual decreases in caloric intake [1]. Selye subsequently showed a similar pattern of reproductive response in restrained rats as those separated from their cage-mates or siblings. These data suggest that similar mechanisms of hypothalamic adaptation on the reproductive system occur in response to exercise training, weight loss, and psychological stress as well as to illness [45]. In Selye’s day, before immunoassays for hormones were available, the level of stress was best indicated by adrenal gland weights. Because reproductive disturbances occurred in parallel, they were also assessed as “adaptive” and related Interstitial atrophy to a generalized stress response. These observations are consistent with current data showing elevated cortisol levels in women with hypothalamic disturbances of ovulation, oligomenorrhea, and amenorrhea [85]. These classical animal stress experiments are only now being reproduced in humans. However, as will be discussed in more detail below, the data available in mature women suggest that a high training intensity and volume is well tolerated if adequate nutrition and a suitable time for adaptation to that exercise are allowed. Finally, although interactions among “treat” or “stress” variables had been postulated in women [6], it has not been experimentally documented. One very important prospective, controlled experimental study in female cynomolgus monkeys has shown that there are synergistic or added reproductive system effects of psychological “stresses” on top of metabolic or energy-­ related threats [86]. 138 Adaptations to Exercise Training Exercise Training Studies in Reproductively Mature Women Only a few studies have prospectively documented changes in cycle and ovulatory characteristics as with exercise training in mature women. The first prospective documentation, in only one woman, used the elasticity of cervical mucus as a marker of the midcycle estradiol peak to show shortening of the luteal phase associated with an increase in weekly running distance [2]. Other early studies showed an increased prevalence of short luteal phase or anovulatory cycles associated with increasing intensity or volume of exercise training [87, 88]. In a group of 14 reproductively mature women (gynecologic age >15 year, mean chronologic age 35 years) who had been training for a marathon, only one-third of a total of 48 cycles prior to a marathon (three cycles/woman) were ovulatory with normal luteal-phase lengths [88]. The only difference between nonovulatory and ovulatory cycles appeared to be the length of the usual training run from approximately 2–5 miles [88]. A study of longer duration (14–15 month) in women not initially proven to be ovulatory showed a decrease in the volume of menstrual blood and lower estradiol levels with marathon training [89]. Running activity increased from 24 to 100 km/week over the study period. Ovulatory characteristics were not examined, however, and the inclusion of participants from ages 24 to 57 years old [89] confounds these outcomes. Nevertheless, in that study, and in none of the others to be subsequently described, did the women develop amenorrhea, despite rapid increases in running activity/intensity mandated by some of the protocols. In Table 8.1 we compare three important prospective studies of exercise and reproduction. These studies have all sought to establish an influence of exercise training on the reproductive hormonal characteristics of both the follicular and the luteal phases of the menstrual cycle as well as ovulatory changes during exercise training: Bullen [27, 90], Bonen [3], and Rogol et al. A. Y. Liu et al. [5]. Because of their importance to this discussion, each study is described in detail below. Bullen and colleagues [27, 90] monitored 28 college-aged women residing at a summer camp by measuring hormonal characteristics for 2 cycles using analysis of daily overnight urines and evening temperatures. These women (whose mean age was 20 years) were confirmed to be ovulatory prior to entry into the study and were also randomly assigned to either weight-loss or weight-maintenance groups. Running activity increased from 4.5 to 10 miles/d by week 5 of the 8-week camp. In addition to running 10 miles/d, women also participated in 3 h/d of varied recreational activities. Bullen and colleagues documented that none of the women in the study developed amenorrhea despite their young age and that they were exposed to several stressors, including change of residence, intense and rapidly increasing exercise training, and caloric restriction (in the weight-loss group). Ovulatory disturbances and shortened luteal-phase cycles were common, however, and only 8 of the 28 women ovulated normally in both cycles. The addition of weight loss to the exercise training caused a further significant increase in ovulation disturbances as well as oligomenorrhea in a few women [27, 90]. Bonen and colleagues [3] set out to determine whether a dose–response between running mileage/week and reproductive function was operative. In particular, by observing sedentary, mature women who ran at varying exercise loads, they tried to determine whether or not a threshold of exercise intensity was present above which luteal-phase disturbances would begin. Bonen [3] monitored mature women over 2–4 month who were variously training at <16, 16–32, or 32–48 km/week. These investigators showed that although there were trends toward shortening of the luteal phase in the first cycle measured after training began, no consistent luteal-phase length changes were documented, nor were there any differences in ovulatory characteristics between women in different intensity groups [3]. A study by Rogol et al. [5] was similar to Bonen’s, but used VO2max testing to document the anaerobic threshold or when lactate began to 8 Exercise and the Hypothalamus: Ovulatory Adaptations 139 Table 8.1 Published prospective studies of exercise training on menstrual cycle and luteal phase lengths Author Bullen et al. [27] Bonen [3] Rogol et al. [5] 28 57 23 Total (n)a Chronologic age 22 (0.6) 30.0 (1.3) 31.4 (1.3) Gynecologic age 10 (0.6) 17.1 (1.4) 17.8 (0.9) Train at lactate threshold <10 miles/week for Mean (SE) Study Exercise + weight maintenance (A) (n = 9) 2 months (A) groups Exercise + weight loss (B) (max of Train above lactate <10 miles/week for −0.45 kg/week) threshold (n = 8) 4 months (B) 10–20 miles/week for 2 month (C) 10–20 miles/week for 4 months (D) 20–30 miles/week for 2 months (E) 20–30 miles/week for 4 months (F) Duration of 2 months 2–4 months 1 year exercise training As described above Start: 6.25 miles/week Exercise Running 4 miles/d progressing to Weeks 1–20: add schedule 10 miles/d by week 5, plus 3.5 h of 1.25 miles every second cycling, tennis, or volleyball week Weeks 20–39: hold at 24 miles/week Weeks 40–end: add 1.25 miles every second week (max of 40 or 65 miles/week) Exercise intensity 70–80% of max aerobic capacity Not reported 6 d/week ran (adjusted each month) at lactate threshold 3 d/week ran at lactate threshold and 3 d/week ran above lactate threshold Sampling method Daily BBT and daily urinary Daily blood samples Daily blood samples day 9 sampling (overnight) through end of cycle Sampling Continuous Every second cycle Every fourth cycle intervals Mean LL not available cycle types Control Mean LL Cycle 1 Mean LL Luteal length during training cycle 14.2 (1.5) 13.9 (0.6) (LL) Mean (SE) Study group A B Run cycle 1 12.6 (1.0) Cycle 4 13.4 (0.7) Cycle 8 13.8 (0.7) %Ovulatory 25 6 Run cycle 3 14.2 (1.5) (only includes groups B, D, and F) %Short luteal 66 63 Cycle 12 12.8 (0.7) phase %Anovulatory 42 81 Detrain, 12.1 (1.3) cycle 3 or 5 NA NA Additional Young gynecologic age stressors Weight loss Away from home Intense exercise training a Number of participants who completed the study 140 be produced. This assessment was used to gradually increase the exercise intensity to maintain physical activity just below or above the “lactate threshold.” This allowed investigators to more accurately document the exercise load, which was gradually increasing over 1 year. Participants’ hormone levels were intensively sampled every 4 months before the next increase in exercise intensity. Rogol et al. [5] also reported that neither running intensity nor duration affected ovulatory function in women training for 1 year at increasing intensities that were maintained either above or below their own adjusted lactate threshold. Several differences exist between the studies of Bullen and those performed by Bonen and Rogol, which at least partially explain their discrepant outcomes. The rapid introduction of a high volume of training and the addition of weight loss in Bullen’s protocol provides a greater stress load and would thus be more likely to lead to ovulatory disturbances than an exercise program alone in older women who remained in their own homes and communities [3, 5]. In addition, the women in Bullen et al.’s [27] study were significantly younger in both chronological and gynecological ages. Another important difference is in design—Bullen and colleagues increased exercise intensity rapidly, whereas the other two studies were more gradual in exercise intensification. Finally, these studies differ in the methods and time course of monitoring. Bullen et al. [27] monitored cycles consecutively and inclusively. In contrast, Bonen and Rogol et al. assessed ovulatory characteristics intermittently every two or every four cycles, respectively. Shortened luteal-phase length or anovulatory cycles may have been missed because monitoring occurred after one or three cycles of probable adaptation to a new exercise load. Any ovulatory disturbances would have likely occurred in the first cycle following the increase in training volume. By the second or fourth cycle after the increase in intensity/ duration of training, adaptation would have occurred, homeostatic balance would be achieved, and normal ovulatory function would have returned. A. Y. Liu et al. We, like Bullen et al. [27], have monitored luteal length and ovulation continuously, but over 1 year in 66 community-dwelling women of varying self-chosen activity levels [4]. As described earlier, all women were confirmed to be normally ovulatory on two consecutive cycles prior to study entry. Despite that, over 80% of the women experienced at least one short luteal phase or anovulatory cycle during the year of study. When the average cycle, luteal phase, and two cycles of hormone levels were used, no differences were found by exercise habit in the number or severity of ovulatory disturbances, or in estradiol and progesterone levels. That was true regardless of whether the women were completing <1 h of aerobic exercise/week (normally active controls), running more than 1 h/week, but not training for a specific event (consistent runners), or runners increasing training in preparation for a marathon that 19 women completed during the study year [4]. The reason for the subclinical ovulatory disturbances that did occur was not initially understood. However, we have subsequently found them to be more prevalent in women scoring high on the Restraint Scale, suggesting they are related to cognitive dietary restraint [69–71]. The same study was recently used to compare the characteristics of the pre-marathon cycle in the marathon-training women with a season-­ matched cycle in the consistent runners. Exercise training without weight loss can be shown to cause shortening of the luteal phase. The luteal-­ phase characteristics of the cycle before the marathon were compared in marathon-training women with their own initial and final cycles and the pre-marathon cycle with a season-matched middle cycle from the consistent runners. Compared to both their own cycles during less intense training and all of the cycles in the consistent runners, significant shortening of the luteal-phase length before the marathon occurred in the marathon-training women (Petit & Prior, Personal communication, 2010). Hypothalamic adaptation to the runners’ baseline exercise probably had occurred before they passed the screening for two consecutive ovulatory cycles and became qualified to enter the study. However, the intensified training before 8 Exercise and the Hypothalamus: Ovulatory Adaptations the marathon appeared to cause shortening of the luteal phase in the cycle prior to the marathon when their training mileage was the greatest. The detailed dietary, weight, body fat, and hormonal characteristics also monitored before the marathon are being studied for explanations other than exercise training to explain the luteal-phase shortening that was documented. These data all suggest that adaptation to increased exercise, even as intense as training for a marathon, normally occurs with only shortening of the luteal phase in well-nourished, reproductively mature women who have no major emotional distress [91]. In addition, as discussed below, adaptation allows a woman’s reproductive system to show rapid shortening of the luteal phase and equally rapid reversion to normal. bservable Changes Prior O to Ovulation Disturbances: Molimina Prior to shortening of the luteal-phase length, which is the first objective change in reproductive function, other observable but even more subtle changes are commonly reported by mature women who are beginning exercise training. The earliest change with moderate, recreational levels of exercise is a decrease in molimina [92] as recorded by the daily Menstrual Cycle Diary© [37] (available at www.cemcor.ca). “Molimina,” whose Greek etymology means “the work of bringing on flow,” includes the set of physical and emotional, but not troublesome, indicators of the coming menstrual flow. Although premenstrual symptoms may occur in both ovulatory and non-ovulatory cycles [93], we previously believed that molimina indicated that ovulation had occurred. However, a recent large study in over 400 unselected women could not confirm the molimina/ovulation association. However, it did show, in the few women who observed it, a highly ovulatory cycle-specific development of axillary breast tenderness during the week before flow [94]. An additional indicator of an ovulatory cycle is the disappearance of elastic or stretchy cervical mucus after the midcycle estrogen surge. Because progesterone inhibits cervical production of elas- 141 tic mucus, this time pattern of the presence and then the disappearance of mucus is also a potential indicator that ovulation has occurred. We asked whether exercise would decrease premenstrual experiences by studying a group of proven ovulatory women runners who were increasing their exercise training over 6 months. Exercise training was associated with decreased fluid symptoms and decreased feelings of depression despite no changes in weight or cycle characteristics [92]. Age and weight matched non-exercising and ovulatory women studied in parallel experienced no significant changes in premenstrual experiences over the same study period [92]. Time Course of Ovulatory Adaptation With the addition of more strenuous training, endocrine changes progress to a shortened luteal phase. The next and more disturbed cycle is anovulatory. This sometimes occurs a straining workload increases [6]. The sedentary woman whose training and cycle characteristics are shown in Fig. 8.6 developed severe back pain during the 12th cycle and did not ovulate. It is likely that she developed anovulation because she not only had to deal with the stress of the pain but also what for her was an important worry that she would be unable to compete in and finish the marathon for which she had trained so hard. In a woman with well-established normally ovulatory cycles (probably after gynecological age 12), exercise-training adaptive changes do not normally progress to anovulation. However, if an additional stressor is added, such as illness, insufficient energy intake, weight loss, and/or emotional stress (see sections “Exercise Training Studies in Reproductively Mature Women” and “Reversibility/Adaptation” in this chapter), anovulation may develop. Amenorrhea will ­usually not develop unless the woman is of young gynecologic age, is not yet sturdily ovulatory, and has stresses in addition to exercise training, such as eating restraint or psychological stress, energy imbalance, rapid induction of exercise, or rapid weight loss. A. Y. Liu et al. 142 Reversibility/Adaptation A few within-person studies are useful to illustrate further the progression and reversibility of ovulatory adaptation. Figures 8.6 and 8.7 show luteal-phase lengths as documented by QBT [91] for 1 year of consecutive cycles in two mature, normal-weight women. One of these women, as discussed above, was a sedentary woman who trained for and ran a marathon during the year of observation (Fig. 8.6). The other was a rather lean and compulsive runner who wanted to become pregnant (Fig. 8.7) [91]. The first woman’s prospective record indicated alternating cycles showing short luteal-phases (<10 d) and normal luteal-phase lengths with anovulation during the cycle before and of the marathon race. As mentioned above, the pain and worry of an injury as well as exercise training likely accounted for anovulatory cycles. A normal luteal-phase length cycle returned when both her emotional stress and her training workload decreased immediately after her successful marathon. Figure 8.7 shows prospective documentation of ovulatory characteristics over 1 year in another woman who was running regularly, but was quite lean and stressed. She showed consistently short luteal-phase cycles early in the year. In an effort to reverse her secondary infertility, she decreased running for one cycle, but this was emotionally stressful. Her secondary infertility was due to inadequate or insufficient luteal-phase characteristics documented by endometrial biopsy. When she stopped running for approximately 6 weeks, she became pregnant. These detailed case histories of two women who monitored their individual exercise and ovulatory characteristics over an extended period Follicular phase Injured Luteal phase Marathon * Anovulatory * 35 30 9 11 11 8 9 12 11 25 7 8 9 13 15 7 Miles run/day 20 5 Days 12 10 3 5 1 9 10 11 12 13 14 1 2 3 4 5 6 7 8 Sequential menstrual cycle number during marathon training Fig. 8.6 This bar graph, illustrating cycle lengths as bars and luteal-phase lengths as blue areas within those bars, shows sequential menstrual cycles and ovulation during 1 year of marathon training in a previously sedentary woman. Note the alternating short and normal luteal phases and progression to anovulatory cycles (in cycles #12 and #13) just after the most intense and highest mile- age of training just before and in the marathon cycle. The arrow shows injury which caused her to decrease training and major emotional stress that likely contributed to the anovulatory cycle. When she decreased her training following the marathon, ovulation and her luteal-phase lengths were restored to normal. (Modified from Ref. [91]) 8 Exercise and the Hypothalamus: Ovulatory Adaptations 143 Follicular phase Endometrial Luteal phase biopsy Pregnancy 30 8 10 8 8 25 9 8 6 8 8 8 20 9 Miles run/day 8 Days 7 15 6 10 8 4 6 4 2 2 0 9 10 11 12 1 2 3 4 5 6 7 8 Sequential menstrual cycle number during marathon training 0 Fig. 8.7 This bar graph is similar in format to Fig. 8.6 and shows the sequential menstrual cycle and ovulatory characteristics in a woman who was training intensely and compulsively. Even with decreased running during the first few cycles, she continued having short lutealphase cycles. Endometrial biopsies (arrows) were consis- tent with luteal-phase deficiency. In the middle of cycle 11, she stopped running and became pregnant before a normal luteal phase could be documented. She carried the child to term and delivered a healthy baby. (Modified from Ref. [91]) of time indicate the rapid hypothalamic adaptation and reversibility of ovulatory disturbances related to exercise training [91]. These data have since been supported in larger samples of women runners [4, 88]. Very few data clearly document adaptation to exercise over longer than a year. As an example, it is useful to observe the second marathon-­ training year in the woman whose cycles before and after her first marathon were documented in Fig. 8.6. Her cycles and ovulatory characteristics before her second marathon a year after her first are shown in Fig. 8.8. During the first marathon, she had shown short luteal-phase cycles progressing to anovulatory cycles the month prior to (M-1) and of (M) the marathon. In her second marathon, 1 year later, luteal length remained normal throughout her training, although her training was similar in volume and intensity to her earlier marathon. It appears possible that by the second year, she had adapted to the marathon training, which allowed her cycles to maintain normal ovulation. Key in each of these stories is the fact that the woman was basically emotionally healthy and maintained normal body weights, and good energy intakes avoiding relative energy deficiency (Fig. 8.4). In mature women, adaptation to exercise training with reversal to normally ovulatory, normal-­ length cycles commonly occurs within one cycle. These adaptive changes of luteal-phase length with increasing exercise training are modeled in Fig. 8.9. Note that, as in the woman described above who trained for her first marathon, by the end of the year, the model suggests that a level of exercise intensity that had provoked ovulatory A. Y. Liu et al. 144 5 0 M-2 Anovulation 10 Anovulation Luteal length (days) 15 M-1 M M+1 Marathon 1 Marathon 2 10 Miles/cycle day Fig. 8.8 Luteal-phase lengths during the cycles before and just after a marathon in the first marathon training and race (as shown in Fig. 8.6) and in subsequent marathon a year later. During the second marathon, despite similar or increased mileage, there were no luteal-phase disturbances documented. This illustrates reproductive adaptation to the levels of exercise this woman was now performing. (Modified from Prior et al. [95]) 5 0 M-2 disturbances now no longer causes a change from a normal ovulatory cycle. Bullen’s study [27] also demonstrated rapid reversibility when training ceased. Although a few women developed oligomenorrhea as well as disturbances of ovulation, all of the women regained both normal cycle intervals and normal ovulatory function within a few months after the end of the summer training camp. Furthermore, it is common for athletic women to become pregnant within months of decreasing their training (and loss of competitive stress), even though they may have had several years of anovulatory cycles or amenorrhea [91, 96, 97]. However, in exercising younger women, in whom the hypothalamus M-1 M M+1 has not fully matured, the return to or achievement of normal ovulatory cycles will often take longer. Although the majority of the data just presented were collected using QBT analyses many years ago, no studies since have closely examined ovulatory characteristics continuously during several months of exercise training. The development of new methods of monitoring ovulation and luteal-phase length, for example, with salivary progesterone [25], should soon allow the nuances of cycle adaptation to be more specifically characterized and mechanisms and modulating factors more carefully delineated. 8 Exercise and the Hypothalamus: Ovulatory Adaptations Vo2 max % Exercise intensity A 80–90 4 hours week 5 80 3 hours week 4 70 2 hours week 60 1 hour week 50 0.5 hour week 0 145 B C E D F 3 2 1 Sedentary 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 3 4 5 6 7 8 9 10 11 Consecutive menstrual cycles 12 13 14 15 Luteal length (days) 16 12 8 4 0 1 2 Fig. 8.9 Theoretical model of the luteal-phase changes that occur over time with increasing exercise in an ovulatory woman who is undergoing exercise training. Note that at the end of the year’s sequence of cycles, despite a considerable exercise load, luteal-phase length and ovulation are normal. (Modified from Prior et al. [95]) Clinical Applications/Treatment If ovulatory disturbances are documented, it is very easy to provide physiological treatment. Evidence suggests and the Centre for Menstrual Cycle and Ovulation Research recommends that persistent ovulatory disturbances should be treated by prescribing either cyclic oral micronized progesterone (300 mg at bedtime) or medroxyprogesterone (10 mg) on days 14–27 of the woman’s own cycle [34, 99] (http://www.cemcor.ca/resources/ cyclic-progesterone-therapy). Although this “treatment” does not directly correct the hypothalamic stressor(s) or energy insufficiency that led to the disturbance in the first place, feedback to the hypothalamus by progesterone may aid in reproductive maturation and ovulatory recovery. The most useful function of cyclic progesterone is to provide physiological levels of progesterone, which will cause regular The practical and clinical implications of ovulatory adaptation to exercise training are not the purpose of this chapter. However, it is important that the clinician and coach be alert to document persistent disturbances of lutealphase length or any anovulatory cycles because we now know they are associated with significant bone loss [98]. These ovulatory disturbances, if observed, are very useful indicators that the exercise training load is excessive for that woman’s level of hypothalamic reproductive maturation and/or when combined with other potentially present stressors, e.g., competitive anxiety, insufficient caloric intake, moving away from home, weight loss, eating restraint, or even illness. A. Y. Liu et al. 146 0.1 Spinal bone density (L1-L4) 1 1 year change (g.cm2- ) DXA DXAe 0.05 0 -0.05 -0.1 A n = 16 B n = 16 C n = 15 D n = 14 Fig. 8.10 This dot-plot figure shows individual rates of 1-year spinal bone mineral density change by dual-energy X-ray absorptiometry (DXA) in 61 active, healthy, normal-­weight women ages 20–35 with amenorrhea, oligomenorrhea, subclinical anovulation, or subclinical short luteal phases stratified by reproductive status and randomized to receive medroxyprogesterone acetate (that acts through the osteoblast progesterone receptor) cyclically for 10 d/cycle (10 mg/d of MPA) with or without active/ calcium therapy (1000 mg/day) or placebo. Women in A were taking cyclic MPA plus calcium; B cyclic MPA plus placebo; C placebo MPA plus calcium; and D double placebos. The effects of cyclic MPA were highly significant (P = 0.0001), and calcium was borderline (P = 0.07). There was a significant 2.0% loss of bone in the double-­ placebo (D) control group [34] menstrual flow if estradiol levels are normal and, acting through the osteoblast progesterone receptor (PR), will increase bone mineral density (BMD) [100]. Cyclic medroxyprogesterone ­(acting through the osteoblast PR) in a randomized, placebo-controlled 1-year trial caused a significant 2% increase in spinal areal BMD in mild-­ moderately active women with hypothalamic disturbances of cycles or ovulation (Fig. 8.10) [34]. Although combined hormonal contraceptives (CHC) are the usual therapy for “functional” hypothalamic amenorrhea or oligomenorrhea, in adolescent women, CHC may cause skeletal [101] and reproductive [102] harm. CHC therapy for treatment of hypothalamic amenorrhea has also been associated with significantly lower rates of recovery (42%) and slower recovery than in women who declined any therapy [103]. The most important reason for the clinician or coach to know about ovulatory disturbances is to recognize them as adaptive and reversible and to teach each woman to observe and understand the menstrual cycle and ovulatory changes she may experience. In this era of “self-help medicine,” keeping the Menstrual Cycle Diary©, QBT (both free at www.cemcor.ca), and training records will increase self-knowledge and thus well-being for health-conscious women. Conclusions This chapter has reviewed the subtle adaptations of women’s reproductive system to gradually and appropriately increasing exercise training. Evidence suggests that decreases in luteal-phase progesterone production and duration (subclinical ovulatory disturbances) are the first and the major adaptive responses of the hypothalamic– pituitary–ovarian system in mature women to increasing exercise intensity. More obvious changes in cycle lengths may occur if the relative energy insufficiency of sport is also present or if 8 Exercise and the Hypothalamus: Ovulatory Adaptations the exercise-training woman is within 5–10 years of menarche. If no additional stressor other than the exercise is present, the subclinical ovulatory disturbances will reverse to normal within the next cycle, even though the exercise training level is maintained. These physiological and psychological changes during exercise training are protective for the individual, are reversible, and cause no long-term harm. However, if subclinical ovulatory disturbances persist (any anovulatory and ≥ 2 short luteal-phase cycles/year) [4], despite clinically normal cycles, bone loss occurs [4, 98]. In addition, fertility is impaired by silent ovulatory disturbances. Persistence of these ovulatory disturbances may be commonly related to the psychological stress caused by cognitive dietary restraint [71] and psychosocial stressors related to women’s inferior cultural status [86]. The benefits of exercise for cardiovascular [104], skeletal [105], and emotional health [106] are well supported by scientific evidence, yet the concept persists that exercise causes women to develop amenorrhea which is an important negative reproductive event. In this chapter, and as described in the concept of relative energy deficiency in sport [61], we have attempted to erase that perception by viewing women’s responses to exercise training as adaptive. When increasing levels of exercise are introduced gradually and energy balance is maintained, adaptation can occur, and the result is a minimal change. Ovulatory disturbances occur normally when initiating a more intense training program or increasing exercise load, but will reverse rapidly to normal once adaptation has occurred. When taken to an extreme or combined with other psychological (including cognitive dietary restraint) or physiological stressors, exercise can cause definite negative reproductive changes. In that circumstance, persistent ovulatory disturbances occur, which, depending on the age, nutritional state, and emotional support of the woman, may progress to oligomenorrhea or amenorrhea. Amenorrhea, although it is uncommonly associated with exercise in mature, ovulatory women, may occur in the face of exercise combined with 147 a negative energy balance or when several stressors coexist, especially in women who have not established regularly ovulatory cycles. Gynecological immaturity is a significant factor impairing the ability of women to appropriately adapt to exercise stress. This implies that caution should be taken in the progression and sequence of increasing exercise intensity in young athletes. In summary, although the concept of adaptation to exercise training has been known for almost 70 years [1] and has been applied to women’s reproduction since the 1980s, few well-­ controlled studies have documented the subtlest evidence of this adaptation: subclinical ovulatory disturbances. References 1. Selye H. The effect of adaptation to various damaging agents on the female sex organs in the rat. Endocrinology. 1939;25:615–24. 2. Shangold MM, Freeman R, Thysen B, Gatz M. The relationship between long-distance running, plasma progesterone, and luteal phase length. Fertil Steril. 1979;31:130–3. 3. Bonen A. Recreational exercise does not impair menstrual cycles: a prospective study. Int J Sports Med. 1992;13:110–20. 4. Prior JC, Vigna YM, Schechter MT, Burgess AE. Spinal bone loss and ovulatory disturbances. New Engl J Med. 1990;323:1221–7. 5. Rogol AD, Weltman A, Weltman JY, Serp RI, Snead DB, Levine S, et al. Durability of the reproductive axis in eumenorrheic women during 1 yr of endurance training. J Appl Physiol. 1992;72:1571. 6. Prior JC. Endocrine “conditioning” with endurance training: a preliminary review. Can J Appl Sport Sci. 1982;7:149–57. 7. Drinkwater BL, Nilson K, Chesnut CH, Bremner WJ, Shainholtz S, Southworth MB. Bone mineral content of amenorrheic and eumenorrheic athletes. New Engl J Med. 1984;311:277–81. 8. Vollman RF. The menstrual cycle. In: Friedman EA, editor. Major problems in obstetrics and gynecology, Vol 7. 1. Toronto: W.B. Saunders Company; 1977. p. 11–193. 9. Prior JC, Naess M, Langhammer A, Forsmo S. Ovulation prevalence in women with spontaneous normal-length menstrual cycles – a population-­ based cohort from HUNT3, Norway. PLOS One. 2015;10(8):e0134473. 10. Abraham GE. The normal menstrual cycle. In: Givens JR, editor. Endocrine causes of menstrual A. Y. Liu et al. 148 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. disorders. 1. Chicago: Year Book Medical Publishers, Inc; 1978. p. 15–44. Prior JC, Vigna YM, Schulzer M, Hall JE, Bonen A. Determination of luteal phase length by quantitative basal temperature methods: validation against the midcycle LH peak. Clin Invest Med. 1990;13:123–31. Taaffe DR, Robinson TR, Snow CM, Marcus R. High-impact exercise promotes bone gain in well-trained female athletes. J Bone Miner Res. 1997;12(2):255–60. Nielsen HK, Brixen K, Bouillon R, Mosekilde L. Changes in biochemical markers of osteoblastic activity during the menstrual cycle. J Clin Endocrinol Metab. 1990;70:1431–7. Prior JC. Perimenopause: the complex endocrinology of the menopausal transition. Endocr Rev. 1998;19:397–428. Welt CK. The physiology and pathophysiology of inhibin, activin and follistatin in female reproduction. Curr Opin Obstet Gynecol. 2002;14(3):317–23. Landgren BH, Unden AL, Diczfalusy E. Hormonal profile of the cycle in 68 normally menstruating women. Acta Endocr Copenhagen. 1980;94:89–98. Fraser IS, Baird DT. Endometrial cystic glandular hyperplasia in adolescent girls. J Obstet Gynecol. 1972;79:1009–13. Van Look PF, Lothian H, Hunter WM, Michie EA, Baird DT. Hypothalamic-pituitary-ovarian function in perimenopausal women. Clin Endocrinol. 1977;7:13–31. Cowan LD, Gordis L, Tonascia JA, Jones GS. Breast cancer incidence in women with a history of progesterone deficiency. Am J Epidemiol. 1981;114(2):209–17. Sherman BM, Korenman SG. Hormonal characteristics of the human menstrual cycle throughout reproductive life. J Clin Investig. 1975;55:699–706. Sowers M, Randolph JF, Crutchfield M, Jannausch ML, Shapiro B, Zhang B, et al. Urinary ovarian and gonadotropin hormone levels in premenopausal women with low bone mass. J Bone Miner Res. 1998;13(7):1191–202. Aksel S, Wiebe RH, Tyson JE, Jones GS. Hormonal findings associated with aluteal cycles. Obstet Gynecol. 1996;48(5):598–602. Soules MR, McLachlan RI, Marit EK, Dahl KD, Cohen NL, Bremner WJ. Luteal phase deficiency: characterization of reproductive hormones over the menstrual cycle. J Clin Endocrinol Metab. 1989;69:804–12. Petsos P, Ratcliffe WA, Heath DF, Anderson DC. Comparison of blood spot, salivary and serum progesterone assays in the normal menstrual cycle. Clin Endocrinol. 1986;24:31–8. Finn MM, Gosling JP, Tallon DF, Madden AT, Meehan FP, Fottrell PF. Normal salivary progesterone levels throughout the ovarian cycle as determined by a direct enzyme immunoassay. Fertil Steril. 1988;50:882–7. 26. Santoro N, Rosenberg J, Adel T, Skurnick JH. Characterization of reproductive hormonal dynamics in the perimenopause. J Clin Endocrinol Metab. 1996;81(4):1495–501. 27. Bullen BA, Skrinar GS, Beitins IZ, von Mering G, Turnbull BA, McArthur JW. Induction of menstrual disorders by strenuous exercise in untrained women. New Engl J Med. 1985;312:1349–53. 28. McNeely MJ, Soules MR. The diagnosis of luteal phase deficiency: a critical review. Fertil Steril. 1988;50:1–15. 29. Hinney B, Henze C, Kuhn W, Wuttke W. The corpus luteum insufficiency: a multifactorial disease. J Clin Endocrinol Metab. 1996;81:565–70. 30. Prior JC, Vigna YM, Barr SI, Kennedy S, Schulzer M, Li DK. Ovulatory premenopausal women lose cancellous spinal bone: a five year prospective study. Bone. 1996;18:261–7. 31. Hitchcock CL, Bishop C, Prior JC, editors. Modelling ovulation and detecting subclinical ovulatory disturbances. Chicago: The 12th conference of the Society for Menstrual Cycle Research; 1997. 32. McCarthy JJ, Rockette HE. A comparison of methods to interpret the basal body temperature graph. Fertil Steril. 1983;39:640–6. 33. Royston JP, Abrams RM. An objective methods for detecting the shift in basal body temperature in women. Biometrics. 1980;36:217–24. 34. Prior JC, Vigna YM, Barr SI, Rexworthy C, Lentle BC. Cyclic medroxyprogesterone treatment increases bone density: a controlled trial in active women with menstrual cycle disturbances. Am J Med. 1994;96:521–30. 35. Bedford JL, Prior JC, Hitchcock CL, Barr SI. Detecting evidence of luteal activity by least-­ squares quantitative basal temperature analysis against urinary progesterone metabolites and the effect of wake-time variability. Eur J Obstet Gynecol Reprod Biol. 2009;146(1):76–80. 36. Prior JC. Physical exercise and the neuroendocrine control of reproduction. Baillieres Clin Endocrinol Metab. 1987;1:299–317. 37. Prior JC. Exercise-associated menstrual disturbances. In: Adashi EY, Rock JA, Rosenwaks Z, editors. Reproductive endocrinology, surgery and technology. New York: Raven Press; 1996. p. 1077–91. 38. Petraglia F, Sutton S, Vale W, Plotsky P. Corticotropin-releasing factor decreases plasma luteinizing hormone levels in female rats by inhibiting gonadotropin-releasing hormone release into hypophysial-portal circulation∗. Endocrinology. 1987;120:1083–8. 39. Hakimi O, Cameron LC. Effect of exercise on ovulation: a systematic review. Sports Med. 2017;47(8):1555–67. 40. Prior JC. Reproduction: exercise-related adaptations and the health of women and men. In: Bouchard C, Shephard RJ, Stephens T, Sutton JR, McPherson BD, 8 Exercise and the Hypothalamus: Ovulatory Adaptations 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. editors. Exercise, fitness and health. 1. Champaign: Human Kineteic Books; 1990. p. 661–75. Barr SI, Janelle KC, Prior JC. Energy intakes are higher during the luteal-phase of ovulatory menstrual cycles. Am J Clin Nutr. 1995;61(1):39–43. Kaplan JR, Adams MR, Clarkson TB, Koritnik DR. Psychological influences on female ‘protection’ among cynomolgus macaques. Atherosclerosis. 1984;53:283–95. Kowalski W, Chatterton RT Jr, Kazer RR, Wentz AC. The impact of subchronic hypercortisolemia on progesterone metabolism and the luteinizing hormone-­ progesterone axis in the Cynomolgus monkey. J Clin Endocrinol Metab. 1993;77(6):1597–603. Chatterton RT, Kazer RR, Rebar RW. Depletion of luteal phase serum progesterone during constant infusion of cortisol phosphate in the cynomolgus monkey ∗†∗Supported by grant HD 21921 from the National Institutes of Health, Bethesda, Maryland. †Presented at the 71st Annual Meeting of the Endocrine Society, Seattle, Washington, June 21, 1989. Fertil Steril. 1991;56(3):547–54. Warren MP, Siris ES, Petrovich C. The influence of severe illness on gonadotropin secretion in the postmenopausal female. J Clin Endocrinol Metab. 1977;45:99–104. Nagata I, Kato K, Seki K, Furuya K. Ovulatory disturbances. Causative factors among Japanese student nurses in a dormitory. J Adolesc Health Care. 1986;7:1–5. Schweiger U, Laessle RG, Schweiger M, Herman F, Riedel W, Pirke KM. Caloric intake, stress and menstrual function in athletes. Fertil Steril. 1988;49:447–50. Warren MP. Effects of undernutrition on reproductive function in the human. Endocr Rev. 1983;4:363–77. Alvero R, Kimzey L, Sebring N, Reynolds J, Loughran M, Nieman L, et al. Effects of fasting on neuroendocrine function and follicle development in lean women. J Clin Endocrinol Metab. 1998;83:76–80. Barron JL, Noakes TD, Levy W, Smith C, Millar RP. Hypothalamic dysfunction in overtrained athletes. J Clin Endocrinol Metab. 1985;60:803–6. Ding JH, Sheckter CB, Drinkwater BL, Soules MR, Bremner WJ. High serum cortisol levels in exercise-associated amenorrhea. Ann Intern Med. 1988;108:530–4. Berga SL, Daniels TL, Giles DE. Women with functional hypothalamic amenorrhea but not other forms of anovulation display amplified cortisol concentrations. Fertil Steril. 1997;67(6):1024–30. Loucks AB, Mortola JF, Girton L, Yen SSC. Alterations in the hypothalamic-pituitary-­ ovarian and the hypothalamic-pituitary-adrenal axes in athletic women. J Clin Endocrinol Metab. 1989;68:402–11. 149 54. Constantini NW, Warren MP. Menstrual dysfunction in swimmers: a distinct entity. J Clin Endocrinol Metab. 1995;80(9):2740–4. 55. Prior JC. Ovulatory disturbances: they do matter. Can J Diagn. 1997;14:64–82. 56. Graham TE, Viswanathan M, Van Dijk JP, Bonen A, George JC. Thermal and metabolic responses to cold by men and by eumenorrheic and amenorrheic women. J Appl Physiol. 1989;67(1):282–90. 57. Loucks AB, Verdun M, Heath EM. Low energy availability, not stress of exercise, alters LH pulsatility in exercising women. J Appl Physiol. 1998;84(1):37–46. 58. Kaye WH, Gwirtsman HE, George DT, Ebert MH, Jimerson DC, Tomai TP, et al. Elevated cerebrospinal fluid levels of immunoreactive corticotropin-­ releasing hormone in anorexia nervosa: relation to state of nutrition, adrenal function, and intensity of depression. J Clin Endocrinol Metab. 1987;64(2):203–8. 59. Pirke KM, Schweiger U, Strowitzki T, Tuschl RJ, Laessle RG, Broocks A, et al. Dieting causes menstrual irregularities in normal weight women through impairment of luteinizing hormone. Fertil Steril. 1989;51:263–8. 60. Suh BY, Liu JH, Berga SL, Quigley ME, Laughlin GA, Yen SSC. Hypercortisolism in patients with functional hypothalamic amenorrhea. J Clin Endocrinol Metab. 1988;66:733–9. 61. Mountjoy M, Sundgot-Borgen J, Burke L, Carter S, Constantini N, Lebrun C, et al. The IOC consensus statement: beyond the female athlete triad--Relative Energy Deficiency in Sport (RED-S). Br J Sports Med. 2014;48(7):491–7. 62. Constantini NW. Medical concerns of the dancer. Book of Abstracts, XXVII FIMS World Congress of Sports Medicine, Budapest, Hungary, 2002;151. 63. Williams NI, Leidy HJ, Hill BR, Lieberman JL, Legro RS, De Souza MJ. Magnitude of daily energy deficit predicts frequency but not severity of menstrual disturbances associated with exercise and caloric restriction. Am J Physiol Endocrinol Metab. 2015;308(1):E29–39. 64. Reed JL, De Souza MJ, Mallinson RJ, Scheid JL, Williams NI. Energy availability discriminates clinical menstrual status in exercising women. J Int Soc Sports Nutr. 2015;12:11. 65. Stunkard AJ, Messick S. The three-factor eating questionnaire to measure dietary restraint, disinhibition and hunger. J Psychosom Res. 1985;29:71–83. 66. Laessle RG, Tuschl RJ, Kotthaus BC, Pirke KM. Behavioral and biological correlates of dietary restraint in normal life. Appetite. 1989;12:83–94. 67. Tuschl RJ, Platte P, Laessle RG, Stichler W, Pirke KM. Energy expenditure and everyday eating behavior in healthy young women. Am J Clin Nutr. 1990;52(1):81–6. 68. Schweiger U, Tuschl RJ, Platte P, Broocks A, Laessle RG, Pirke KM. Everyday eating behavior A. Y. Liu et al. 150 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. and menstrual function in young women. Fertil Steril. 1992;57:771–5. Barr SI, Prior JC, Vigna YM. Restrained eating and ovulatory disturbances: possible implications for bone health. Am J Clin Nutr. 1994;59:92–7. Barr SI, Janelle KC, Prior JC. Vegetarian versus nonvegetarian diets, dietary restraint, and subclinical ovulatory disturbances: prospective six month study. Am J Clin Nutr. 1994;60:887–94. Bedford JL, Prior JC, Barr SI. A prospective exploration of cognitive dietary restraint, subclinical ovulatory disturbances, cortisol and change in bone density over two years in healthy young women. J Clin Endocrinol Metab. 2010;95(7):3291–9. Allaway HC, Southmayd EA, De Souza MJ. The physiology of functional hypothalamic amenorrhea associated with energy deficiency in exercising women and in women with anorexia nervosa. Horm Mol Biol Clin Invest. 2016;25(2):91–119. Sathi P, Kalyan S, Hitchcock CL, Pudek M, Prior JC. Progesterone Therapy increases Free Thyroxine Levels- data from a randomized placebo-controlled 12-week hot flush trial. Clin Endocrinol (Oxf). 2013;79:282–7. Bonen A, Belcastro AN, Simpson AA. Profiles of menstrual cycle hormones in teenage athletes. J Appl Physiol. 1981;50:545–51. Ronkainen HR. Depressed follicle stimulating hormone, luteinizing hormone releasing hormone, thyrotopin releasing hormone, and metoclopramide. Fertil Steril. 1985;40:755–9. Malina RM. Menarche in athletes: a synthesis and hypothesis. Ann Hum Biol. 1983;10:1–24. Rees M. Menarche when and why? Lancet. 1993;342(8884):1375–6. Warren MP. The effects of exercise on pubertal progression and reproductive function in girls. J Clin Endocrinol Metab. 1980;51(5):1150–7. Treloar SA, Martin NG. Age at menarche as a fitness trait: nonadditive genetic variance detected in a large twin sample. Am J Hum Genet. 1990;47(1):137–48. Frisch RE, Gotz-Welbergen AV, McArthur JW, Albright TE, Witschi J, Bullen BA, et al. Delayed menarche and amenorrhea of college athletes in relation to age of onset of training. J Am Med Assoc. 1981;246:1559–63. McEwen BS. Protective and damaging effects of stress mediators. Seminars in Medicine of the Beth Israel Deaconess Medical Center. New Engl J Med. 1998;338(3):171–9. Prior JC, Vigna YM, Watson D. Spironolactone with physiological female gonadal steroids in the presurgical therapy of male to female transexuals: a new observation. Arch Sex Behav. 1989;18:49–57. Barr SI, Petit MA, Vigna YM, Prior JC. Eating attitudes and habitual calcium intake in peripubertal girls are associated with initial bone mineral content and its change over 2 years. J Bone Miner Res. 2001;16:940–7. 84. Rivier C, Rivier JE, Vale W. Stress-induced inhibition of reproductive functions: role of endogenous corticotropin-­ releasing factor. Science. 1986;231:607–9. 85. Berga SL, Loucks-Daniels TL, Adler LJ, Chrousos GP, Cameron JL, Matthews KA, et al. Cerebrospinal fluid levels of corticotropin-releasing hormone in women with functional hypothalamic amenorrhea. Am J Obstet Gynecol. 2000;182(4):776–81. 86. Williams NI, Berga SL, Cameron JL. Synergism between psychosocial and metabolic stressors: impact on reproductive function in cynomolgus monkeys. Am J Physiol Endocrinol Metab. 2007;293(1):E270–E6. 87. Bonen A, Keizer HA. Athletic menstrual cycle irregularity: endocrine response to exercise and training. Phys Sportsmed. 1984;12:78–94. 88. Prior JC, Cameron K, Ho Yeun B, Thomas J. Menstrual cycle changes with marathon training: anovulation and short luteal phase. Can J Appl Sport Sci. 1982;7:173–7. 89. Boyden TW, Pamenter RW, Stanforth PR, Rotkis TC, Wilmore JH. Sex steroids and endurance running in women. Fertil Steril. 1983;39:629–32. 90. Beitins IZ, McArthur JW, Turnbull BA, Skrinar GS, Bullen BA. Exercise induces two types of human luteal dysfunction: confirmation by urinary free progesterone. J Clin Endocrinol Metab. 1991;72:1350–8. 91. Prior JC, Ho Yeun B, Clement P, Bowie L, Thomas J. Reversible luteal phase changes and infertility associated with marathon training. Lancet. 1982;1:269–70. 92. Prior JC, Vigna YM, Alojado N, Sciarretta D, Schulzer M. Conditioning exercise decreases premenstrual symptoms: a prospective controlled six month trial. Fertil Steril. 1987;47:402–8. 93. Prior JC. Premenstrual symptoms and signs. In: Rabel RE, Bope ET, editors. Conn’s current therapy 2002. New York: W.B. Saunders Company; 2002. p. 1078–80. 94. Prior JC, Konishi C, Hitchcock CL, Kingwell E, Janssen P, Cheung AP, et al. Does molimina indicate ovulation? Prospective data in a hormonally documented single-cycle in spontaneously menstruating women. Int J Environ Res Pub Health. 2018;15(5):1016. 95. Prior JC. Luteal phase defects and anovulation: adaptive alterations occurring with conditioning exercise. Semin Reprod Endocrinol. 1985;3:27–33. 96. Bonen A. Exercise-induced menstrual cycle changes a functional, temporary adaptation to metabolic stress. Sports Med. 1994;17(6):373–92. 97. Cohen G, Prior JC, Vigna YM, Pride SM. Intense training during the first two trimesters of unapparent pregnancy. Phys Sportsmed. 1989;17:11–7. 98. Li D, Hitchcock CL, Barr SI, Yu T, Prior JC. Negative spinal bone mineral density changes 8 Exercise and the Hypothalamus: Ovulatory Adaptations and subclinical ovulatory disturbances--prospective data in healthy premenopausal women with regular menstrual cycles. Epidemiol Rev. 2014; 36(137):147. 99. Prior JC. Ovulatory disturbances and amenorrhea: a physiologic approach to diagnosis and therapy. In: Rosenberg JA, editor. Women’s health in primary care. Baltimore: Williams & Wilkins; 1997. p. 437–52. 100. Prior JC. Progesterone for the prevention and treatment of osteoporosis in women. Climacteric. 2018;21:366–74. 101. Goshtasebi A, Subotic Brajic T, Scholes D, Beres Lederer Goldberg T, Berenson A, Prior JC. Adolescent use of combined hormonal contraception and peak bone mineral density accrual: a meta-analysis of international prospective controlled studies. Clin Endocrinol. 2019;90(4):517–24. 151 102. Prior JC. Adolescents’ use of combined hormonal contraceptives for menstrual cycle-related problem treatment and contraception: evidence of potential lifelong negative reproductive and bone effects. Women’s Reprod Health. 2016;3(2):73–92. 103. Falsetti L, Gambera A, Barbetti L, Specchia C. Long-­ term follow-up of functional hypothalamic amenorrhea and prognostic factors. J Clin Endocrinol Metab. 2002;87(2):500–5. 104. Powell KE, Thompson PD, Caspersen CJ, Kendrick JS. Physical activity and the incidence of coronary heart disease. Ann Rev Public Health. 1987;8:253–87. 105. Petit MA, Prior JC, Barr SI. Running and ovulation positively change cancellous bone in premenopausal women. Med Sci Sports Exerc. 1999;31(6):780–7. 106. McCann L, Holmes DS. Influence of aerobic exercise on depression. J Pers Soc Psychol. 1984;46:1142–7. 9 Adrenergic Regulation of Energy Metabolism Michael Kjær and Kai Lange Introduction During exercise, energy turnover increases and adrenergic mechanisms play an important role in this regulation. In addition, increased adrenergic activity during exercise also results in an increased heart rate and in an enhanced force of myocardial contraction as well as in vasoconstriction in the splanchnic circulation, in the kidneys, and in noncontracting muscles. These circulatory changes favor a redistribution of blood flow to exercising muscle as well as an increased cardiac output [1]. Furthermore, the adrenergic activity stimulates sweat glands and thereby influences thermoregulation, and it causes an increased contractility of skeletal muscle as well as influences exercise-induced suppression of components of the human immune system. In the present chapter, it is demonstrated how adrenergic activity can influence substrate mobilization and utilization both directly and indirectly via secretion of hormones. M. Kjær (*) · K. Lange Department of Clinical Medicine, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark e-mail: michaelkjaer@sund.ku.dk drenergic Responses to Acute A Exercise Adrenergic activity can be assessed both by direct measurements of electrical activity in superficial sympathetic nerves and by measurement of circulating norepinephrine and epinephrine in the blood. The direct recording of sympathetic activity can be performed to resting muscle only, but during exercise of, e.g., the arms, sympathetic activity to the resting leg muscle has been shown to increase with progressively increasing intensity of arm exercise [2]. In addition to these measurements, a correlation has been found between sympathetic nerve activity and plasma levels of norepinephrine [3]. Although a correlation between circulating norepinephrine and direct recordings of sympathetic nerve activity from the peroneal nerve has been demonstrated during exercise, the increase in sympathetic outflow to the various regions of the body differs somewhat during exercise. During exercise, using methods to measure norepinephrine spillover, it has been demonstrated that the increase in sympathetic activity during exercise is dominated by an increased sympathetic activity directed toward active muscle. During two-­ legged exercise, approx. 50% of all circulating norepinephrine is released from sympathetic nerve endings in active muscle. Furthermore, when arm exercise is added to leg exercise, the norepinephrine © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_9 153 154 spillover from active leg muscle also increases despite unchanged work output and unchanged blood flow to the leg muscles [4]. In addition to norepinephrine released from sympathetic nerve endings, epinephrine is released from the adrenal medulla in response to sympathetic neural activity during exercise. The circulating epinephrine is responsible for the major adrenergic effect on energy metabolism during exercise compared with norepinephrine. In the present chapter, the adrenergic effect on carbohydrate and fat metabolism will be discussed, but epinephrine per se has been shown also to increase protein metabolism in isolated electrically stimulated rat muscle [5]. The levels of circulating free norepinephrine and epinephrine increase with exercise intensity expressed by the percentage of maximal individual performance (%VO2 max). This holds true both during prolonged exercise and in response to short-term intermittent exercise and to intense weight training. The increase in plasma norepinephrine and epinephrine occurs rapidly in arterial blood, and it has been calculated that the half-life of epinephrine is around 2–3 min during exercise. Circulating levels of catecholamines can only be considered as overall markers of sympathoadrenergic activity and are influenced not only by secretion but also by clearance of the hormone. Whereas clearance of norepinephrine is difficult to determine on a whole-body level owing to the fact that it is extracted at two levels in series, namely, both the lung and the systemic organs [6], the turnover of epinephrine can be studied in humans, using a radio-labeled tracer. It has been shown that whole-body clearance of epinephrine increases by 15% at low exercise intensities and decreases around 20% below basal levels after more intense exercise [7]. However, since the increase in plasma epinephrine seen during dynamic exercise in humans is five- to tenfold, these changes are caused by increases in secretion from the adrenal medulla rather than by changes in clearance. Among the major contributors to epinephrine clearance are the hepatosplanchnic area and the kidneys. M. Kjær and K. Lange otor Control and Reflex Influence M on Adrenergic Response In experiments using partial neuromuscular blockade to weaken the muscle force and thereby increase the motor center activity needed to produce a certain force output, it was found that exercise-induced increases in levels of circulating catecholamines were augmented compared to control experiments with saline infusion [8]. These findings are supported by experiments in paralyzed cats where direct stimulation of the subthalamic locomotor areas in the brain resulted in adrenergic hormonal responses similar to the ones seen during voluntary exercise [9]. Together, these experiments support the view that motor center activity can directly stimulate sympathoadrenergic activity during exercise directly and independently of feedback from contracting muscle. That central factors linked to exercise intensity are not sufficient to elicit a maximal adrenergic response can be demonstrated in different ways. When exercising a small muscle group (e.g., one knee extensor) even at maximal intensity, only a small catecholamine response can be observed [4]. Furthermore, when maximal work output was reduced by more than 60% with a neuromuscular blockade (tubocurarine), despite subjects working at the highest possible effort, adrenergic responses were far from maximal [10]. In addition to central factors, peripheral neural feedback can be demonstrated using lumbar epidural anesthesia in doses sufficiently high to block impulses in thin afferent nerves but preserving motor nerves and the ability to perform exercise to the highest possible degree. During static exercise, but not during dynamic exercise, catecholamine responses were inhibited when afferent responses were absent [11, 12]. Interestingly, both ACTH and β-endorphin responses during submaximal exercise were abolished during epidural anesthesia [11, 13]. In support of a role of afferent nerves in adrenergic responses, plasma catecholamines increased in response to direct stimulation of these nerve fibers in cats [14]. An alternative model to study feedback mechanisms during exercise is to use 9 Adrenergic Regulation of Energy Metabolism patients with metabolic deficiencies. Both in myophosphorylase (McArdle’s disease) and phosphofructokinase deficiency and in mitochondrial myopathy, an excessive neuroendocrine response and exaggerated mobilization of extramuscular substrate (glucose and free fatty acid (FFA)) were found, most likely a coupling toward the oxidative demands of the muscle cell rather than to the oxidative capacity of the working muscle [15–17]. drenergic Activity After Physical A Training Vigorous endurance training will reduce the catecholamine response to a given absolute workload [18], whereas neither sympathetic nerve activity nor norepinephrine levels at maximal workloads differ between individuals with different training status [19]. This supports the view that physical training does not alter the capacity of the sympathetic nervous system, but that responses to submaximal exercise are linked closely to the relative rather than to the absolute workload [20]. Surprisingly, however, it has in a 24-h study been found that highly trained individuals had a higher catecholamine release over the day compared with sedentary individuals [21]. Epinephrine response in trained individuals vs. sedentary has been shown to be enlarged when stimulated by a variety of stimuli, such as hypoglycemia, caffeine, glucagon, hypoxia, and hypercapnia [20, 22–25]. This indicates that the capacity to secrete epinephrine from the adrenal medulla improves with training. In rats that underwent 10 weeks of intense swim training, the adrenal medullary volume and the adrenal content of epinephrine were larger in trained rats compared with controls who were either weight matched, sham-trained, or cold-stressed [26]. Although these findings indicate that the improved secretion capacity of epinephrine is a result of training, this will most likely require several years of training. In well-trained athletes who underwent hypoglycemia before and 4–5 weeks after an injury that resulted in inactiv- 155 ity, epinephrine responses did not change with this short-lasting alteration in activity level [27]. However, still it is interesting that endocrine glands apparently are able to adapt to physical training and alter their secretion capacity, similar to other tissues like muscle and heart. Hepato-splanchnic Glucose Production and Adrenergic Activity During intense exercise the rise in hepatic glucose production was parallel with a rise in plasma catecholamine levels [28–30]. In addition, in models where electrically induced cycling was used in spinal cord-injured individuals with impaired sympathoadrenergic activity, hepatic glucose production was abolished [31]. In swimming rats, the removal of the adrenal medulla reduced the hepatic glycogenolysis [32], as well as the exercise-induced increase in hepatic glucose production in running rats [33]. However, most studies have been unable to demonstrate any effect of epinephrine on liver glycogen breakdown during exercise [34–37]. In running dogs, evidence has been provided that epinephrine may play a minor role in liver glucose output late during exercise [38] probably owing to an increased gluconeogenic precursor level. Furthermore, adrenalectomized individuals maintain a normal rise in hepatic glucose production during exercise [39], and only when epinephrine is infused in these patients, hepatic glucose production was augmented during the early stages of exercise (unpublished observation). Direct stimulation of liver nerves caused an increase in hepatic glycogenolysis, and the hypothesis has been put forward that liver nerves are important for the exercise-induced rise in liver glucose output. In contrast to this, surgical or chemical denervation of the liver in various species did not reduce the exercise-induced increment in hepatic glucose production [32, 33, 40, 41], which indicates that sympathetic liver nerves are not essential during exercise. In humans, the role of liver nerves and epinephrine has been 156 studied with application of local anesthesia around the sympathetic celiac ganglion innervating the liver, pancreas, and adrenal medulla [42]. Pancreatic hormones were standardized by infusion of somatostatin, glucagon, and insulin. During blockade, the exercise-induced epinephrine response was inhibited by up to 90%, and presumably liver nerves were also blocked, but this did not diminish the glucose production response to exercise. This indicates that sympathoadrenergic activity is not responsible for an exercise-induced rise in splanchnic glucose output. In further support of this hypothesis, the exercise-induced increase in liver glucose production was identical in liver-transplanted patients compared to healthy control subjects as well as in kidney-transplanted patients who received a similar hormonal and immunosuppressive drug treatment as liver-transplanted patients [43]. Liver-transplanted patients were investigated approx. 8 months after surgery, and no sign of reinnervation occurred in any of the patients as judged by the content of norepinephrine in liver biopsies [44]. Finally, in recent experiments in exercising dogs that underwent a selective blockade of hepatic α- and β-receptors, it was demonstrated that circulating norepinephrine and epinephrine do not participate in the stimulation of glucose production during intense exercise [45, 46]. Taken together, sympathetic liver nerves or circulating norepinephrine play no role in glucose mobilization from the liver during exercise, and circulating epinephrine only plays a minor role during intense exercise and late during prolonged exercise. drenergic Effect on Skeletal A Muscle Carbohydrate Metabolism Muscle contractions per se increase glucose uptake, and humoral factors can modify this [47]. Insulin and contractions have a synergistic effect on glucose uptake with contractions [48], whereas epinephrine has been demonstrated to decrease glucose clearance in running dogs [49]. In addition to this, femoral arterial infusion of epinephrine into an exercising leg in humans caused a M. Kjær and K. Lange reduction in the normal exercise-induced glucose uptake [50]. More recently, it has been shown that in adrenalectomized individuals performing leg cycling for 45 min at 50% VO2 max followed by 15 min at 85% VO2 max, the rise in glucose uptake during exercise was reduced when epinephrine was infused to substitute plasma epinephrine levels normally observed during exercise (unpublished observation). The mechanism behind this is at present unknown but could be related to an enhanced glycogenolysis, increased intramuscular glucose concentration, or altered uptake of FFA, all changes that can influence glucose uptake. It has been shown that adrenergic activity can enhance the glycogen breakdown in muscle during contraction both in exercising animals [51] and in humans [50, 52]. However, those studies often used supraphysiological doses of epinephrine, and later studies in humans using lower doses have only been able to demonstrate a higher activation of phosphorylase, but could not demonstrate any marked increase in glycogen breakdown [53]. Noradrenergic activity probably does not play any role in muscle glycogenolysis, since unilateral hind limb sympathectomy did not diminish glycogen breakdown in swimming rats [54]. Sympathoadrenergic Activity and Fat Metabolism Lipolysis in fat tissue is enhanced by β-adrenergic activity, and catecholamine responsiveness of β-adrenergic receptors in adipose tissue is increased after acute exercise [55]. By the use of microdialysis of subcutaneous abdominal tissue, it was demonstrated that nonselective β-adrenoceptor blockade inhibited the exercise-­ induced increase in dialysate levels of glycerol [56]. Although this indicates a role for adrenergic activity in fat metabolism during exercise, the relative role between sympathetic nerve activity and circulating norepinephrine/epinephrine is currently not known. Intravenous infusion of epinephrine in resting humans caused an increase in lipolytic activity as determined by microdialysis 9 Adrenergic Regulation of Energy Metabolism of subcutaneous adipose tissue, an effect that was desensitized by repeated epinephrine infusions [57]. The direct role of sympathetic nerve activity on adipose tissue has recently been addressed using microdialysis, and it was found that during handgrip exercise, the increase in umbilical glycerol release was attenuated in spinal cord-injured individuals with impaired sympathetic nerve activity when compared with healthy control individuals [58]. It should be noted that this very moderate type of stress was not able to document any increase in lipolysis in the clavicular region. Furthermore, in a recent study, glycerol output in subcutaneous abdominal adipose tissue was found to be lower during prolonged arm-­cranking in spinal cord-injured individuals compared with controls performing a similar relative workload (unpublished observation). Taken together, indices are provided that sympathetic nerves to adipose tissue stimulate lipolysis directly during exercise. If regional differences (visceral vs. subcutaneous fat) exist in responsiveness of the adipose tissue toward increased sympathetic activity, this could play an important role in the treatment of adipositas. Not only adipose tissue but also intramuscular fat can be stimulated by catecholamines, and both lipoprotein lipase (LPL) and hormone-­ sensitive lipase (HSL) play important roles in this regulation [59]. HSL might be under control by both contractions and epinephrine, and it has recently been shown that activation of HSL and glycogen phosphorylase occurs in parallel in adrenalectomized individuals who receive infusion with epinephrine during exercise (unpublished observation). This could indicate that mobilization of intramuscular triglyceride and glycogen occurs simultaneously, stimulated by adrenergic activity, and that choice of substrate for energy production takes place at another level. Summary Physical exercise causes an increase in adrenergic activity that can be determined both by changes in plasma catecholamines and in intraneural sympathetic activity. Release of norepinephrine from 157 contracting muscles and release of epinephrine from the adrenal medulla are major contributors to high levels of plasma catecholamines. Both feed-forward stimulation from motor centers in the brain and afferent impulses from working muscles stimulate sympathoadrenergic activity, and a coupling to oxidative demands of the working muscle is likely. Long-­term physical training increases the size and secretory capacity of the adrenal medulla, which may improve exercise capacity. Sympathoadrenergic activity only plays a minor role in regulation of hepatic glucose release, but via depressing insulin secretion and influencing target tissue, adrenergic activity improves glycogen and fatty acid mobilization. References 1. Rowell LR. Human circulation regulation during physical stress. New York: Oxford University Press; 1986. 2. Victor R, Seals DR, Mark AL. Differential control of heart rate and sympathetic nerve activity during dynamic exercise: insight from direct intraneural recordings in humans. J Clin Invest. 1987;79:508–16. 3. Searls DR, Victor RG, Mark AL. Plasma norepinephrine and muscle sympathetic discharge during rhythmic exercise in humans. J Appl Physiol. 1988;65:940–4. 4. Savard G, Richter EA, Strange S, Kiens B, Christensen NJ, Saltin B. Norepinephrine spillover from skeletal muscle during exercise in humans: role of muscle mass. Am J Phys. 1989;257:H1812–8. 5. Nie ZT, Lisjo S, Åstrand PO, Henriksson J. In-vitro stimulation of the rat epitrochlearis muscle II. Effects of catecholamines and nutrients on protein degradation and amino acid metabolism. Acta Physiol Scand. 1989;135:523–9. 6. Esler M, Jennings G, Korner P, Blomberry P, Sacharias N, Leonard P. Measurement of total and organ-­specific norepinephrine kinetics in humans. Am J Phys. 1984;247:E21–8. 7. Kjær M, Christensen NJ, Sonne B, Richter EA, Galbo H. Effect of exercise on epinephrine turnover in trained and untrained male subjects. J Appl Physiol. 1985;59:1061–7. 8. Kjær M, Secher NH, Bach FW, Galbo H. Role of motor center activity for hormonal changes and substrate mobilization in exercising man. Am J Phys. 1987;253:R687–95. 9. Vissing J, Iwamoto GA, Rybicki KJ, Galbo H, Mitchell JH. Mobilization of glucoregulatory hormones and glucose by hypothalamic locomotor centers. Am J Phys. 1989;257:E722–8. 158 10. Galbo H, Kjær M, Secher NH. Cardiovascular, ventilatory and catecholamine responses to maximal dynamic exercise in partially curarized man. J Physiol. 1987;389:557–68. 11. Kjær M, Secher NH, Bach FW, Sheikh S, Galbo H. Hormonal and metabolic responses to exercise in humans: effect of sensory nervous blockade. Am J Phys. 1989;257:E95–101. 12. Kjær M, Secher NH, Bach FW, Galbo H, Reeves DR, Mitchell JH. Hormonal, metabolic and cardiovascular responses to static exercise in man: influence of epidural anesthesia. Am J Phys. 1991;261:214–20. 13. Klokker M, Kjær M, Secher NH, Hanel B, Worm L, Kappel M, et al. Natural killer cell response to exercise in humans: effect of hypoxia and epidural anesthesia. J Appl Physiol. 1995;78:709–16. 14. Vissing J, Iwamoto GA, Fuchs IE, Galbo H, Mitchell JH. Reflex control of glucoregulatory exercise responses by group III and IV muscle afferents. Am J Phys. 1994;266:R824–30. 15. Vissing J, Lewis SF, Galbo H, Haller RG. Effect of deficient muscular glycogenolysis on extramuscular fuel production in exercise. J Appl Physiol. 1992;72:1773–9. 16. Vissing J, Galbo H, Haller R. Paradoxically enhanced glucose production during exercise in humans with blocked glycolysis due to muscle phosphofructokinase deficiency. Neurology. 1996;47:766–71. 17. Vissing J, Galbo H, Haller RG. Exercise fuel mobilization in mitochondrial myopathy: a metabolic dilemma. Ann Neurol. 1996;40:655–62. 18. Winder WW, Hagberg JM, Hickson RC, Ehsani AA, McLane JA. Time course of sympathoadrenergic adaptation to endurance exercise training in man. J Appl Physiol. 1978;45:370–4. 19. Svedenhag J. The sympathoadrenal system in physical conditioning. Acta Physiol Scand. 1985;125(Suppl 543):1–74. 20. Kjær M, Bangsbo J, Lortie G, Galbo H. Hormonal response to exercise in man: influence of hypoxia and physical training. Am J Phys. 1988;254:R197–203. 21. Dela F, Mikines KJ, Linstow M, Galbo H. Heart rate and plasma catecholamines during 24 hour everyday life in trained and untrained men. J Appl Physiol. 1992;73:2389–95. 22. Kjær M, Mikines KJ, Christensen NJ, Tronier B, Vinten J, Sonne B, et al. Glucose turnover and hormonal changes during insulin-induced hypoglycemia in trained humans. J Appl Physiol. 1984;57:21–7. 23. Kjær M, Farrel PA, Christensen NJ, Galbo H. Increased epinephrine response and inaccurate glucoregulation in exercising athletes. J Appl Physiol. 1986;61:1693–700. 24. Kjær M, Galbo H. The effect of physical training on the capacity to secrete epinephrine. J Appl Physiol. 1988;64:11–6. 25. LeBlanc J, Jobin M, Cote J, Samson P, Labri A. Enhanced metabolic response to caffeine in exercise-trained human subjects. J Appl Physiol. 1985;59:832–7. M. Kjær and K. Lange 26. Stallknecht B, Kjær M, Ploug T, Maroun L, Ohkuwa T, Vinten J, et al. Diminished epinephrine response to hypoglycemia despite enlarged adrenal medulla in trained rats. Am J Phys. 1990;259:R998–1003. 27. Kjær M, Mikines KJ, Linstow M, Nicolaisen T, Galbo H. Effect of 5 weeks detraining on epinephrine response to insulin induced hypoglycemia in athletes. J Appl Physiol. 1992;72:1201–5. 28. Kjær M, Kiens B, Hargreaves M, Richter EA. Influence of active muscle mass on glucose homeostasis during exercise in humans. J Appl Physiol. 1991;71:552–7. 29. Marliss EB, Simantirakis E, Miles PDG, Purnon C, Gougeon R, Field CJ, et al. Glucoregulatory and hormonal responses to repeated bouts of intense exercise in normal male subjects. J Appl Physiol. 1991;71:924–33. 30. Sigal R, Fisher SF, Halter JB, Vranic M, Marliss EB. The roles of catecholamines in glucoregulation in intense exercise as defined by the islet cell clamp technique. Diabetes. 1996;45:148–56. 31. Kjær M, Pollack SF, Mohr T, Weiss H, Gleim GW, Bach FW, et al. Regulation of glucose turnover and hormonal responses during exercise: electrical induced cycling in tetraplegic humans. Am J Phys. 1996;271:R191–9. 32. Richter EA, Galbo H, Holst JJ, Sonne B. Significance of glucagon for insulin secretion and hepatic glycogenolysis during exercise in rats. Horm Metab Res. 1981;13:323–6. 33. Sonne B, Mikines KJ, Richter EA, Christensen NJ, Galbo H. Role of liver nerves and adrenal medulla in glucose turnover of running rats. J Appl Physiol. 1985;59:1640–6. 34. Arnall DA, Marker JC, Conlee RK, Winder WW. Effect of infusing epinephrine on liver and muscle glycogenolysis during exercise in rats. Am J Phys. 1986;250:E641–9. 35. Carlson KI, Marker JC, Arnall DA, Terry ML, Yang HT, Lindsay LG, et al. Epinephrine is unessential for stimulation of liver glycogenolysis during exercise. J Appl Physiol. 1985;58:544–8. 36. Marker JC, Arnall DA, Conlee RK, Winder WW. Effect of adrenodemedullation on metabolic responses to high intensity exercise. Am J Phys. 1986;251:R552–9. 37. Winder WW, Arogyasami J, Yang HT, Thompson KG, Nelson A, Kelly KP, et al. Effects of glucose infusion in exercising rats. J Appl Physiol. 1988;64:2300–5. 38. Moates JM, Lacy DB, Goldstein RE, Cherrington AD, Wasserman DH. The metabolic role of the exercise induced increment in epinephrine in the dog. Am J Phys. 1988;255:E428–36. 39. Hoelzer DR, Dalsky GP, Schwartz NS, Clutter WE, Shah SD, Holloszy JO, et al. Epinephrine is not critical to prevention of hypoglycemia during exercise in humans. Am J Phys. 1986;251:E104–10. 40. Wasserman DH, Williams PE, Lacy DB, Bracy D, Cherrington AD. Hepatic nerves are not essential to the increase in hepatic glucose production during muscular work. Am J Phys. 1990;259:E195–203. 9 Adrenergic Regulation of Energy Metabolism 41. Wasserman DH, Cherrington AD. Regulation of extramuscular fuel sources during exercise. In: Rowell LB, Shepherd JT, editors. Handbook of physiology. Columbia: Bermedica Production; 1996. p. 1036–74. 42. Kjær M, Engfred K, Fernandes A, Secher NH, Galbo H. Regulation of hepatic glucose production during exercise in man: role of sympathoadrenergic activity. Am J Phys. 1993;265:E275–83. 43. Kjær M, Keiding S, Engfred K, Rasmussen K, Sonne B, Kirkegård P, et al. Glucose homeostasis during exercise in humans with a liver or kidney transplant. Am J Phys. 1995;268:E636–44. 44. Kjær M, Jurlander J, Keiding S, Galbo H, Kirkegaard P, Hage E. No reinnervation of hepatic sympathetic nerves after liver transplantation in human subjects. J Hepatol. 1994;20:97–100. 45. Coker RH, Krishna MG, Brooks Lacy D, Allen EJ, Wasserman DH. Sympathetic drive to liver and nonhepatic splanchnic tissue during heavy exercise. J Appl Physiol. 1997;82:1244–9. 46. Coker RH, Krishna MG, Brooks Lacy D, Bracy DP, Wasserman DH. Role of hepatic alpha- and beta-adrenergic receptor stimulation on hepatic glucose production during heavy exercise. Am J Phys. 1997;273:E831–8. 47. Richter EA. Glucose utilization. In: Rowell LB, Shepherd JT, editors. Handbook of physiology 1997. Columbia: Bermedica Production; 1996. p. 912–51. 48. Ploug T, Galbo H, Richter EA. Increased muscle glucose uptake during contractions: no need for insulin. Am J Phys. 1984;247:E726–31. 49. Issekutz B. Effect of epinephrine on carbohydrate metabolism in exercising dogs. Metabolism. 1985;34:457–64. 50. Jansson E, Hjemdahl P, Kaijser L. Epinephrine-­ induced changes in muscle carbohydrate metabolism 159 51. 52. 53. 54. 55. 56. 57. 58. 59. during exercise in male subjects. J Appl Physiol. 1986;60:1466–70. Richter EA, Ruderman NB, Gavras H, Belur ER, Galbo H. Muscle glycogenolysis during exercise: dual control by epinephrine and contractions. Am J Phys. 1982;242:E25–32. Spriet LL, Ren JM, Hultman E. Epinephrine infusion enhances glycogenolysis during prolonged electrical stimulation. J Appl Physiol. 1988;64:1439–44. Chesley A, Hultman E, Spriet LL. Effects of epinephrine infusion on muscle glycogenolysis during intense aerobic exercise. Am J Phys. 1995;268:E127–34. Richter EA, Galbo H, Christensen NJ. Control of exercise induced muscular glycogenolysis by adrenal medullary hormones in rats. J Appl Physiol. 1981;50:21–6. Wahrenberg H, Engfeldt P, Bolinder J, Arner P. Acute adaptation in adrenergic control of lipolysis during physical exercise in humans. Am J Phys. 1987;253:E383–90. Arner P, Kriegholm E, Engfeldt P, Bolinder J. Adrenergic regulation of lipolysis in situ at rest and during exercise. J Clin Invest. 1990;85:893–8. Stallknecht B, Bülow J, Frandsen E, Galbo H. Desensitization of human adipose tissue to adrenaline stimulation studied by microdialysis. J Physiol. 1997;500:271–82. Karlsson AK, Elam M, Friberg P, Biering-Sørensen F, Sullivan L, Lønnroth P. Regulation of lipolysis by the sympathetic nervous system: a microdialysis study in normal and spinal cord injured subjects. Metabolism. 1997;46:388–94. Oscai LB, Essig DA, Palmer WK. Lipase regulation of muscle triglyceride hydrolysis. J Appl Physiol. 1990;69:1571–7. Sex Differences in Energy Balance and Weight Control 10 Kristin S. Ondrak I ntroduction: What Is Energy Balance and What Factors Influence It? Before understanding how men and women differ with regard to the storage and utilization of energy, it is important to define the concept of energy balance. Historically, physiologists described energy balance as the difference between the amount of kcal ingested through food vs. the amount of kcal expended through physical activity and basal metabolic processes. The resulting value is one’s body mass. With this balance in mind, when caloric intake is similar to caloric expenditure, a state of neutral energy balance occurs and body mass remains stable. However, when intake exceeds expenditure, positive energy balance ensues and the body stores the excess energy, thereby increasing body mass. The opposite results when expenditure exceeds intake, and this is termed negative energy balance or energy deficit [38]. While these relationships are well supported in research, it is also important to note that a variety of factors impact this relationship, and one cannot simply compare caloric deficits or excesses over the short term and expect body mass to change accordingly [25]. Some of these factors include hormones, age, acute bouts of exercise, chronic exercise K. S. Ondrak (*) Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC, USA e-mail: kondrak@unc.edu training, and changes in body composition, namely, gains or losses in lean mass. These factors will be discussed throughout this chapter, as well as how they differ between men and women. In addition to recognizing that energy balance is more complex than it may appear, it is important to keep in mind that intake and expenditure should be compared over the long term. When even small deficits or excesses in daily energy balance occur day after day, substantial changes in body mass can result. For example, in a 4-year cohort analysis study of women, researchers found that the addition of daily sugar-sweetened beverages to one’s diet resulted in substantial increases in body mass and increased their risk for type II diabetes [41]. These researchers found that by increasing the consumption of sugar-­ sweetened beverages from less than one per week to one or more per day, these women consumed an extra 358 kcal/day and gained ∼4.5 kg on average [41]. Thus, seemingly small additions to one’s daily diet can quickly add up to substantial changes in body mass and energy balance. Along the same lines, small changes in daily habits may result in large increases in calories expended. For example, standing still, as in an elevator, is associated with a caloric expenditure of 1.3 METS, while taking the stairs expends 4.0–8.8 METS depending on the speed [15]. Another component to discuss is the expenditure of calories and how that impacts energy balance. When individuals are compared, there is a © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_10 161 K. S. Ondrak 162 substantial amount of variation in the number of calories burned in a given day. This daily energy expenditure (EE) is comprised of three main components: resting metabolic rate (RMR) which comprises 60–75% of the total calories expended, diet-induced thermogenesis which accounts for 10–15% of the total, and activity thermogenesis, which is subdivided into exercise and non-­ exercise components [17]. The largest component, RMR, is closely related to the amount of fat-free mass (i.e., lean mass) an individual has, and it is generally higher in men compared to women [37]. That is, men tend to burn more calories even at rest compared to women, and this is largely attributed to their higher amounts of metabolically active lean mass. Another complicating factor is age; research has shown that basal metabolic rate decreases as we get older [24]. For example, BMR was 4.6% lower in older participants, compared to younger, in a study of individuals ranging from 15 to 64 years of age [28]. Much of what we know about age-related declines in BMR stems from cross-sectional studies, though, with most of the difference being attributed to declines in lean mass. Additional longitudinal research on age-related declines in BMR is necessary [24]. In addition to its role in elevating RMR, lean body mass is also related to diet-induced thermogenesis. That is, greater amounts of lean mass are associated with a greater number of calories being burned following consumption of food. Other factors that influence diet-induced thermogenesis include age, sex, fitness level, and menstrual cycle phase. Interestingly, some studies have shown that body composition and physical activity levels were more closely related to EE than were age and sex [28]. In fact, fat-free mass had the strongest relationship with 24-h EE, and the values were similar between women and men (r2 = 0.79 for females and 0.76 for males) [28]. As might be expected, there is wide variation in the number of kilocalories an individual burns each day via physical activity. Daily physical activity levels and related caloric expenditure from activity thermogenesis generally decline with age in both men and women [17]. Research on sex differences in physical activity levels is mixed, however; some researchers have reported lower levels of physical activity EE in women compared to men [37], while others have reported no difference [17]. Similarly, sex differences in measures of EE disappear after taking body composition into account (EE per kg fat-free mass). In summary, these sex differences in RMR, diet-­ induced thermogenesis, and activity thermogenesis begin to explain how men and women differ with regard to overall energy balance and weight control. After reviewing the components of daily EE and related energy balance, one can see that there are many factors involved in energy balance and the propensity to change or maintain body mass. This chapter will examine the relationship between these variables with an emphasis on the roles of hormones in weight control and how they differ in males and females. It is important to note that this chapter focuses on healthy, normal-­ weight adults as the norm, as these processes differ in adults who are overweight or obese as well as in children. ormones That Impact Energy H Balance, Distribution of Fat, and Weight Control Numerous hormones influence EE and body mass in humans. Some of the same hormones influence the distribution of body fat, and not surprisingly, the patterns of storage differ between women and men. For simplicity in this chapter, the hormones are grouped according to their primary functions as follows: metabolic (leptin, insulin, ghrelin, anorexigens, and orexigens), sex (estrogen and androgens), and stress (catecholamines and cortisol) hormones. Their roles and impact on body mass and weight control are explained in each respective subsection. Metabolic Hormones Several hormones with metabolic functions impact energy balance in humans, namely, leptin, insulin, and ghrelin. Leptin, the most recently discovered hormone, is catabolic in nature and provides satiety signals to the brain [16, 19, 42, 10 Sex Differences in Energy Balance and Weight Control 50]. It is released by adipose tissue, and its circulating levels are closely related to the amount of fat mass in adults. Leptin is released in greater quantities from subcutaneous fat stores compared to visceral locations [13]. Some researchers suggest that leptin is more closely related to total body fat levels in females compared to males [51]. It follows that these authors reported that females are more sensitive to the actions of leptin than are males. Leptin plays an important role in regulating long-term energy balance rather than the acute fluctuations that occur after each meal [21]. Once released, leptin, along with insulin, acts at the level of the hypothalamus where it induces feelings of fullness, signaling for the person to stop eating. However, leptin’s role in energy balance is complex as it is influenced by numerous other hormones including thyroid hormones (T4 and T3), cortisol, insulin, and growth hormone (GH) [35]. Insulin is another metabolic hormone that influences energy balance. It is secreted from pancreatic β-cells in response to increases in blood glucose. Insulin levels are indicative of visceral fat levels in humans [8, 12]. The correlation between body fat and insulin is particularly strong in males, and some authors suggest that men are more sensitive to insulin than females [51]. Thus, these sex-related differences in insulin and leptin sensitivity provide a possible mechanism explaining the metabolic differences in weight control among men and women. Similar to leptin, insulin reduces appetite over the long term [16, 19, 21]. As a result, these hormones are often classified as anorexigenic (i.e., appetite suppressants). Ironically, individuals with excess body mass (i.e., overweight or obese) often display resistance to leptin and/or insulin [16, 19, 33]. This suggests that being in a state of chronic positive energy balance alters the body’s ability to respond to satiety cues and regulate blood glucose levels. These changes also explain why overweight and obese men and women are at an increased risk for developing impaired glucose tolerance and subsequently type II diabetes. Ghrelin is another metabolic hormone impacting energy balance. It stimulates hunger in the short term and, not surprisingly, is released in 163 great amounts by the stomach [21, 29, 30, 53]. Ghrelin levels fall after meals in a manner proportional to the energy load of the meal consumed, suggesting that this hormone plays a role in inducing satiety and regulating energy balance [21, 29]. Interestingly, ghrelin’s actions are opposite of insulin, although ghrelin plays a role in its release [21]. Insulin and ghrelin are negatively correlated; individuals with high insulin levels tend to have low ghrelin levels. It is not surprising that this hormonal profile of elevated insulin and low ghrelin is common in overweight and obese individuals in particular, as ghrelin levels and body mass index (BMI) are inversely correlated [21, 34]. Ghrelin’s regulation of acute energy balance in the short term is due to its effect on the hypothalamus as it stimulates the release of numerous signals that increase hunger. These orexigens (appetite stimulants) include neuropeptide Y (NPY), agouti-related protein (AgRP), and melanocyte-­stimulating hormone (α-MSH) [21, 29]. In one of the first studies of ghrelin during exercise, plasma acylated ghrelin levels and related ratings of hunger declined during and following an acute running bout in young men [9]. This highlights ghrelin’s role in stimulating appetite and is intuitive that ghrelin levels are suppressed during exercise. Notably, in a well-designed study of male and female twin pairs, resting plasma ghrelin levels were significantly higher in women compared to men [34]. Taken together, these sex differences in anorexigens and orexigens suggest that the signals controlling hunger and satiety differ among men and women. Additional anorexigens including cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide YY (PYY) are discussed in the next subsection along with the influence of sex hormones on each. These differences are another important consideration for understanding sex-related differences in energy balance and weight control. ex Hormones, Orexigens, S and Anorexigens Estrogens and androgens play a large role in body weight regulation, fat distribution, and energy balance in humans and rodent models. Of these sex K. S. Ondrak 164 hormones, women tend to have higher levels of circulating estrogen (technically ­ estrogens – estradiol-β17, estrone, and estriol), while men display greater levels of androgens. Estrogen is related to decreased levels of visceral fat in men and women; alternatively, androgens are related to lower levels of visceral fat in males but higher levels of visceral fat in females [3, 5, 8]. In addition to estrogen’s role guiding the development of secondary sex characteristics and bone mass, estrogen also has important metabolic roles including the reduction of appetite and body mass [1]. Additionally, estrogen interacts with the metabolic hormones leptin and insulin to influence body fat distribution and overall energy balance. The release of estrogen impacts appetite by also decreasing the action and/or effectiveness of several orexigens (i.e., appetite stimulants) including ghrelin, neuropeptide Y (NPY), and melanin-concentrating hormone (MCH) [36, 47]. This data supports estrogen’s role in decreasing food intake through its influence on orexigens. Estrogen also leads to a reduction in food intake through its effects on anorexigenic hormones including insulin, leptin, serotonin, and cholecystokinin (CCK) [11, 14]. It is important to recognize the importance of other appetite suppressants Fig. 10.1 Theoretical model of estrogen’s relationship with fat, leptin, and insulin such as cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide YY (PYY) [44]. Along with leptin, insulin, and ghrelin, these factors have been shown to decrease hunger signals at hypothalamus [44]. Hormonal Interactions Researchers have developed a potential model to explain the relationship between estrogen, fat distribution, and leptin and insulin [43], as shown in Fig. 10.1. They theorize that in premenopausal women, estrogen reduces visceral fat through enhanced lipolysis and decreased lipogenesis. Estrogen also retains subcutaneous fat and is related to increases in resting leptin and reductions in resting insulin levels. However, in men and postmenopausal women, these researchers propose that the lower levels of estrogen and lowered activity of estrogen receptor alpha are related to increases in visceral fat and reductions in subcutaneous fat and leptin, while concomitantly insulin levels are increased [31, 43]. Some of this may be explained by the direct relationship between leptin and subcutaneous fat as the latter secretes leptin. These sexrelated differences in hormone levels and fat distribution have been supported by other researchers as well [10]. Normal Estrogen Levels (As in Premenopausal Women) ↓ Visceral fat (↑ lipolysis, ↓ lipogenesis) retains subcutaneous fat ↑ leptin, ↓ insulin Lower Estrogen Levels (As in Postmenopausal Women and Men) ↑ visceral fat ↓ subcutaneous fat ↓ leptin, ↑ insulin 10 Sex Differences in Energy Balance and Weight Control Androgens such as testosterone and dehydroepiandrosterone (DHEA; and its sulfated form, DHEA-S) also play an important role in energy balance, specifically in influencing where males and females store their body fat. Women tend to deposit and retain more adipose tissue around their hips, buttocks, and thighs, known as a “gynoid” or “pear” body shape. On the other hand, men tend to store more fat around their waist and midsection, known as an “android” or “apple” body shape [5]. The underlying cause of the gynoid shape in women and android shape in men may be due to differences in adipogenesis and the environment (e.g., hormonal milieu) within developing adipose cells. For example, research has shown that women have greater levels of early-differentiated adipocytes compared to men (measured in abdominal and femoral fat depots) [45]. These authors also speculated that sex differences in regional fat distribution may be due to differences in the microenvironment of the cells and related apoptosis, innervation, blood supply, and responsiveness to hormones [45]. In addition to the aforementioned sex differences in body fat distribution, men tend to have higher levels of visceral fat, while women generally store more fat subcutaneously. Androgen levels may play a role in these relationships as greater amounts of visceral fat have been associated with lower androgen levels in men and excess androgen levels in women [5]. Unfortunately for men, visceral fat carries an increased cardiovascular disease risk compared to subcutaneous fat. This often puts men at an increased risk for cardiovascular disease [3–5]. Not surprisingly, inverse correlations have been reported between body fat and EE from physical activity in men (r = −0.34, p < 0.03) [37]. Therefore, sex differences in estrogen and androgens are related to body fat and its distribution; these differences in turn influence EE and balance in men and women. Stress Hormones Stress hormones such as catecholamines and cortisol are another group of hormones that have a large impact on energy balance. To further com- 165 plicate the matter, stress hormones also interact with sex hormones, thus altering their actions [32]. Catecholamines such as epinephrine and norepinephrine are released in response to sympathetic nervous system stimulation when a stressor occurs, whether real or perceived; a common example is exercise. In response to catecholamine release, appetite centers in the hypothalamus are suppressed, and related food intake declines. The primary function of catecholamines and the stress hormone cortisol is to provide energy for the body to face the stressor. Rather than stimulating appetite, these hormones cause the body to break down stored energy, and one example is by stimulating lipolysis. This process is also enhanced by thyroid hormones, cortisol, growth hormone, and estrogen [35]. Thus, there are numerous hormonal signals triggering fat breakdown throughout the body. These hormones and their related lipolytic actions are extremely important during exercise, especially at low to moderate intensities. Catecholamines also increase available energy by increasing glycogenolysis in both the liver and the muscle [54]. The data concerning sex differences in catecholamines at rest and during exercise is conflicting; some studies have shown no difference in men and women, while others have reported slightly higher levels of epinephrine and norepinephrine in men [54]. Likewise, research has shown that men and women have similar levels of both blood and salivary cortisol measures at rest [27]. However, these authors identified sex differences in salivary cortisol in response to stress, such that women in the luteal phase of their menstrual cycle had similar responses to men and both were greater than women in the follicular phase of their menstrual cycle or women on oral contraceptives [27]. This suggests that both sex and the menstrual cycle phase of women should be considered when evaluating cortisol levels and their impact on energy balance. While the functions of numerous metabolic, sex, and stress hormones that impact energy balance were discussed, the following sections will describe how these hormones are affected by physical activity. The related changes in appetite, 166 energy intake, and energy balance will also be discussed, and sex-related differences in these relationships will be discussed when possible. ow Does Physical Activity H Influence Appetite, Satiety, and Energy Balance? It is commonly believed that increased level of physical activity and exercise leads to stimulation of appetite. However, research in this area has reported mixed results. For example, researchers have shown that in general, physical activity does not have a large influence on the balance between intake and expenditure [7]. That is, increases in physical activity do not necessarily stimulate appetite, just as reductions in physical activity do not lead to substantial decreases in appetite. Blundell [6] has built upon the work of previous researchers and proposed two zones to describe how changes in physical activity relate to changes in appetite and food intake, a regulated zone and a non-regulated zone. In the regulated zone, increases in physical activity are related to increased drive to eat; however in the non-­regulated zone, reductions in physical activity, or becoming sedentary, are not indicative of reductions in food intake. Thus, lack of regulation shows that caloric expenditure is not always related to hunger signals. This author also suggested that increasing one’s level of physical activity should help move them into the regulated zone where these variables are more tightly connected, hence regulated. These trends between physical activity level and energy intake were supported in a recent review of cross-sectional research [2]. These authors reported that low levels of habitual physical activity were associated with higher levels of energy intake compared to those with medium and even high levels of physical activity; however, individuals reporting very high levels of physical activity had the highest level of energy intake as one would expect [2]. Hormonal changes also exist in relation to low energy intake. For example, ghrelin, cortisol, and NPY have been shown to increase, while leptin and K. S. Ondrak PYY, among others, decrease in women experiencing chronic negative energy balance [20]. The relationship between an acute bout of exercise and appetite was summarized recently in a comprehensive review by Dorling et al. [18]. Following acute bouts of aerobic exercise, studies have shown a slight suppression of appetite, especially when the bout was ≥60% of VO2 peak, and no consistent sex differences have been shown [18]. This suppression is likely related to reductions in acylated ghrelin which is reduced following exercise of this intensity [18]. These alterations do not last long, though, with hormonal levels and related appetite returning to normal levels within hours of the cessation of exercise. The effect of chronic exercise training on appetite and food intake is not as clear, as studies have shown increases, no change, and even reductions in appetite [18]. The next question, then, is whether sex differences exist in the relationship between physical activity and appetite. The literature in this area is mixed. In a recent review article, Thackray et al. [46] concluded that there was little to no evidence showing sex differences in the relationship between these variables. However, other researchers have reported that women exhibit a greater tendency to either increase energy intake following physical activity or have a more difficult time achieving negative caloric balance and weight loss via physical activity, compared to men [7]. This conclusion has been supported by other researchers as well [23, 48] and by the greater prevalence of obesity in women worldwide compared to men (15% of women vs. 11% of men) [52]. Similar results were reported in a review of 290 participants from 22 studies as physical activity was inversely related to percent body fat in males (partial r = 0.35, p < 0.001) but not in females (partial r = 0.16, p > 0.05), after accounting for age [48]. While the mechanisms behind these differences were beyond the scope of the reviews, authors have hypothesized that sex differences in fat may be attributable to women’s need for sufficient fat stores for successful reproduction [7, 23]. In another study of exercise and appetite, a group of 12 normal-weight men and women 10 Sex Differences in Energy Balance and Weight Control exercised for 14 days, and the resulting changes in energy balance were examined [49]. Participants took part in periods of no additional exercise as well as moderate- and high-intensity exercise, with the order counterbalanced, and were fed ad libitum. The authors reported that the additional EE from the exercise did not elicit equal increases in energy intake; rather the average caloric compensation was only ∼30%. This yielded average negative energy balances ranging from −0.9 to −3.8 MJ/day in women and −1.6 to −4.7 MJ/day in men [49]. The authors acknowledge that while tightly controlled, this study only represents the initial compensation to exercise-­induced energy deficits and longer studies are needed to elucidate the chronic relationships between these variables. Such studies as these provide additional data that contributes to our understanding of sex-related differences in weight control and long-term energy balance. However, much more work in this area of research is necessary. 167 resting leptin and insulin and increases in acylated ghrelin in response to exercise performed in a state of negative energy balance, and women had higher acylated ghrelin and lower insulin following the bout, compared to men [23]. This supports the notion that following physical activity, women’s appetite is stimulated to a greater degree than men’s [23]. These researchers proposed a model to help explain some trends they observed. Specifically, they proposed that in men, physical activity reduces appetite but does not change metabolic hormones such as ghrelin, insulin, and leptin substantially. Therefore, there would be no compensatory changes in energy intake for men, and the energy deficit caused by the increased expenditure would result in reduced body fat. Conversely, in women, physical activity may have no effect on appetite yet cause large hormonal changes. These changes, along with the maintenance of appetite in women, may result in a state of positive energy balance which may preserve or even increase their levels of body fat [23]. This model may explain why women tend to maintain or even gain body mass or fat in Sex Differences in Exercise-Induced response to physical activity, whereas men typically do not. As previously described, the comHormonal Changes mon theory explaining these sex differences is Energy expenditure from physical activity is that the hormonal differences exist to protect influenced by the metabolic-, stress-, and sex-­ women’s fat mass to a greater extent than men’s related hormones described earlier in this chap- in order to ensure successful reproduction. ter. In turn, hormone release is altered in response However, not all studies have supported these sex to physical activity and exercise. Acute bouts of differences in the hormones regulating appetite exercise are related to increases in catechol- or their response to exercise [46]. Nonetheless, amines, growth hormone, cortisol, thyroid hor- the theoretical model of Hagobian and Braun is mones, estrogen, and androgens, while insulin an interesting hypothesis that future research and leptin tend to decrease [35]. These patterns of needs to examine more closely. hormonal release differ somewhat in response to Researchers have also examined sex differchronic exercise training, as many are decreased ences in substrate utilization during exercise. In a in response, but some remain unchanged. study of seven men and seven women endurance-­ To further complicate these relationships, trained cyclists matched by peak oxygen uptake some authors have found that hormonal changes (VO2 peak) per kg lean body mass, there were no in response to exercise may differ between men sex differences in respiratory exchange ratio and women. For example, GH levels have been (RER) during moderate-intensity exercise, shown to increase to a greater degree in women, ­indicating similar contributions from fat and carcompared to men, during exercise [39]. bohydrates [40]. Likewise, there were no sex difAdditionally, in a study examining the hormonal ferences in insulin, epinephrine, or norepinephrine changes following exercise performed in several concentrations during exercise. However, the energy states, researchers noted reductions in sources of fat differed between the sexes as men K. S. Ondrak 168 derived less energy from myocellular triacylglycerols compared to females, and males also had a larger greater proportion of energy that was unaccounted for in fat and carbohydrates sources. Other researchers have reported conflicting results regarding sex differences in fuel metabolism during cycling. Some researchers found that women rely more heavily on fats during exercise (51% vs. 44% for women and men, respectively), while men obtain more energy from carbohydrates (53% and 46%, respectively) when cycling for 2 hours at 40% of their maximal oxygen uptake (VO2 max) [26]. Furthermore, these exercise responses are affected in women by the phase of the menstrual cycle and associated estrogen hormonal changes [22]. These differences are likely attributable to the higher concentrations of epinephrine and norepinephrine seen in men compared to women. It is important to recognize that while conflicting results are often reported, readers must consider the intensity and duration of the exercise within studies as they have a large influence on substrate use and the related hormone response. In summary, collectively these studies provide additional data supporting sex-related differences in energy usage, energy balance, and ultimately weight control. Summary This chapter examined the influence of several hormones and their effect on energy balance and weight control in men and women. Sex differences were explored and include the following: females generally store more fat subcutaneously, while men store more fat in visceral locations; females are more sensitive to the actions of leptin, while men are more sensitive to insulin; and each of these hormones is closely related to one’s level of adiposity. These sex differences often place men at an increased risk for the development of cardiovascular disease due to its association with visceral fat. In addition, the hormonal profile of women favors conservation of fat mass, most likely for reproduction, and some data show that women respond differently to physical activity compared to men. These differences suggest that sex-specific strategies may be necessary for maintaining or altering energy balance and body mass in men and women. References 1. Asarian L, Geary N. Modulation of appetite by gonadal steroid hormones. Philoso Trans R Soc Lond B Biol Sci. 2006;361:1251–63. 2. Beaulieu K, Hopkins M, Blundell J, Finlayson G. Does habitual physical activity increase the sensitivity of the appetite control system? A systematic review. Sports Med. 2016;46:1897–919. 3. Bjorntorp P. Abdominal fat distribution and the metabolic syndrome. J Cardiovasc Pharmacol. 1992;20(Suppl 8):S26–8. 4. Bjorntorp P. Body fat distribution, insulin resistance, and metabolic diseases. Nutrition. 1997;13:795–803. 5. Blouin K, Boivin A, Tchernof A. Androgens and body fat distribution. J Steroid Biochem Mol Biol. 2007;108:272–80. 6. Blundell JE. Physical activity and appetite control: can we close the energy gap? Nutr Bull. 2011;36:356–66. 7. Blundell JE, King NA. Exercise, appetite control, and energy balance. Nutrition. 2000;16(7/8):519–22. 8. Bouchard C, Despres JP, Mauriege P. Genetic and nongenetic determinants of regional fat distribution. Endocr Rev. 1993;14:72–93. 9. Broom DR, Stensel DJ, Bishop NC, Burns SF, Miyashita M. Exercised-induced suppression of acylated ghrelin in humans. J Appl Physiol. 2007;102:2165–71. 10. Brown LM, Clegg DJ. Central effects of estradiol in the regulation of food intake, body weight, and adiposity. J Steroid Biochem Mol Biol. 2010;122:65–73. 11. Butera PC, Bradway DM, Cataldo NJ. Modulation of the satiety effect of cholecystokinin by estradiol. Physiol Behav. 1993;53(6):1235–8. 12. Carey DG, Jenkins AB, Campbell LV, Freund J, Chisholm DJ. Abdominal fat and insulin resistance in normal and overweight women: direct measurements reveal a strong relationship in subjects at both low and high risk of NIDDM. Diabetes. 1996;45:633–8. 13. Casabiell X, Pineiro V, Peino R, Lage M, Camina J, et al. Gender differences in both spontaneous and stimulated leptin secretion by human omental adipose tissue in vitro: dexamethasone and estradiol stimulate leptin release in women, but not in men. J Clin Endocrinol Metabol. 1998;83:2149–55. 14. Clegg DJ, Brown LM, Woods SC, Benoit SC. Gonadal hormones determine sensitivity to central leptin and insulin. Diabetes. 2006;55(4):978–87. 15. Compendium of Physical Activities. Supported by Arizona State University and the National Cancer Institute. Accessed at https://sites.google.com/site/ compendiumofphysicalactivities/home on May 14, 2019. 10 Sex Differences in Energy Balance and Weight Control 16. Davis JF, Choi DL, Benoit SC. Insulin, leptin and reward. Trends Endocrinol Metab. 2010;21(2):1–12. 17. Donahoo WT, Levine JA, Melanson EL. Variability in energy expenditure and its components. Curr Opin Clin Nutr Metab Care. 2004;7:599–605. 18. Dorling J, Broom DR, Burns SF, Clayton DJ, Deighton K, James LJ, King JA, Miyashita M, Thackray AE, Batterham RL, Stensel DJ. Acute and chronic effects of exercise on appetite, energy intake, and appetite-­ related hormones: the modulating effect of adiposity, sex, and habitual physical activity. Nutrients. 2018;10:1140. 19. Friedman JF, Halaas JL. Leptin and the regulation of body weight in mammals. Nature. 1998;395:763–70. 20. Fuqua JS, Rogol AD. Neuroendocrine alterations in the exercising human: implications for energy homeostasis. Metabolim. 2013;62(7):911–21. 21. Gil-Campos M, Aguilera CM, Canete R, Gil A. Ghrelin: a hormone regulating food intake and energy homeostasis. Br J Nutr. 2006;96:201–26. 22. Hackney AC, Curley CS, Nicklas BJ. Physiological responses to submaximal exercise at the mid-­follicular ovulatory and mid-luteal phases of the menstrual cycle. Scand J Med Sci Sports. 1991;1:94–8. 23. Hagobian TA, Braun B. Physical activity and hormonal regulation of appetite: sex differences and weight control. Exerc Sport Sci Rev. 2010;38(1):25–30. 24. Henry CJK. Mechanisms of changes in basal metabolism during ageing. Eur J Clin Nutr. 2000;54(Suppl 3):S77–91. 25. Hopkins M, Blundell JE. Energy balance, body composition, sedentariness and appetite regulation: pathways to obesity. Clin Sci. 2016;130:1615–28. 26. Horton TJ, Pagliassotti MJ, Hobbs K, Hill JO. Fuel metabolism in men and women during and after long-duration exercise. J Appl Physiol. 1998;85(5):1823–32. 27. Kirschbaum C, Kudielka BM, Gaab J, Schommer NC, Hellhammer DH. Impact of gender, menstrual cycle phase, and oral contraceptives on the activity of the hypothalamus-pituitary-adrenal axis. Psychosom Med. 1999;61(2):154–62. 28. Klausen B, Toubro S, Astrup A. Age and sex effects on energy expenditure. Am J Clin Nutr. 1997;65:895–907. 29. Kojima M, Kangawa K. Ghrelin: structure and function. Physiol Rev. 2005;85(2):495–522. 30. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-­ releasing acylated peptide from stomach. Nature. 1999;402(6762):656–60. 31. Krotkiewski M, Bjorntorp P, Sjostrom L, Smith U. Impact of obesity on metabolism in men and women. Importance of regional adipose tissue distribution. J Clin Invest. 1983;72(3):1150–62. 32. Lovejoy JC, Sainsbury A, Stock Conference 2008 Working Group. Sex differences in obesity and the regulation of energy homeostasis. Obes Rev. 2009;10:154–67. 169 33. Lustig RH, Sen S, Soberman JE, Velasquez-Mieyer PA. Obesity, leptin resistance, and the effects of insulin reduction. Int J Obes. 2004;28:1344–8. 34. Makovey J, Naganathan V, Seibel M, Sambrook P. Gender differences in plasma ghrelin and its relation to body composition and bone – an opposite sex twin study. Clin Endocrinol. 2007;66:530–7. 35. McMurray RG, Hackney AC. Interactions of metabolic hormones, adipose tissue and exercise. Sports Med. 2005;35(5):393–412. 36. Messina MM, Boersma G, Overton JM, Eckel LA. Estradiol decreases the orexigenic effect of melanin-­ concentrating hormones in ovariectomized rats. Physiol Behav. 2006;88(4–5):523–8. 37. Paul DR, Novotny JA, Rumpler WV. Effects of the interaction of sex and food intake on the relation between energy expenditure and body composition. Am J Clin Nutr. 2004;79:385–9. 38. Prentice A, Jebb S. Energy intake/physical activity interactions in the homeostasis of body weight regulation. Nutr Rev. 2004;62(7):S98–104. 39. Pritzlaff-Roy CJ, Wideman L, Weltman J, Abbott R, Gutgesell M, Hartman ML, et al. Gender governs the relationship between exercise intensity and growth hormone release in young adults. J Appl Physiol. 2002;92:2053–60. 40. Roepstorff C, Steffensen CH, Madsen M, Stallknecht B, Kanstrup IL, Richter EA, et al. Gender differences in substrate utilization during submaximal exercise in endurance-trained subjects. Am J Physiol Endocrinol Metab. 2002;282:E435–47. 41. Schulze MB, Manson JE, Ludwig DS, Colditz GA, Stampfer MJ, Willett WC, et al. Sugar-sweetened beverages, weight gain, and incidence of type 2 diabetes in young and middle-aged women. J Am Med Assoc. 2004;292:927–34. 42. Schwartz MW, Woods SC, Porte DJ, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000;404:661–71. 43. Shi H, Seeley RJ, Clegg DJ. Sexual differences in the control of energy homeostasis. Front Neuroendocrinol. 2009;30:396–404. 44. Stensel D. Exercise, appetite and appetite-regulating hormones: implications for food intake and weight control. Ann Nutr Metab. 2010;57(supp 2):36–42. 45. Tchoukalova YD, Koutari C, Votruba SB, Tchkonia T, Giorgadze N, Thomou T, et al. Sex- and depot-­ dependent differences in adipogenesis in normal weight humans. Obesity. 2010;18(10):1875–80. 46. Thackray AE, Deighton K, King JA, Stensel DJ. Exercise, appetite and weight control: Are there differences between men and women? Nutrients. 2016;8(9):583. 47. Wade GN, Gray JM, Bartness TJ. Gonadal influences on adiposity. Int J Obes. 1985;9(Supp. 1):83–92. 48. Westerterp KR, Goran MI. Relationship between physical activity related energy expenditure and body composition: a gender difference. Int J Obes. 1997;21:184–8. 170 49. Whybrow S, Hughes DA, Ritz P, Johnstone AM, Horgan GW, King N, et al. The effect of an incremental increase in exercise on appetite, eating behavior and energy balance in lean men and women feeding ad libitum. Br J Nutr. 2008;100:1109–15. 50. Woods SC, Schwartz MW, Baskin DG, Seeley RJ. Food intake and the regulation of body weight. Annu Rev Psychol. 2000;51:255–77. 51. Woods SC, Gotoh K, Clegg DJ. Gender differences in the control of energy homeostasis. Exp Biol Med. 2003;228(10):1175–80. K. S. Ondrak 52. World Health Organization, Obesity and overweight. https://www.who.int/en/news-room/fact-sheets/ detail/obesity-and-overweight. Accessed 16 Dec 2018. 53. Wren AM, Seal LJ, Cohen MA, Brynes AE, Frost GS, Murphy KG, et al. Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab. 2001;86(12):5992–5. 54. Zouhal H, Jacob C, Delamarche P, Gratas-Delamarche A. Catecholamines and the effects of exercise, training and gender. Sports Med. 2008;38(5):401–23. Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function 11 Anne B. Loucks Abbreviations ACSM BMI BW EA EI FFM GH GnRH HPG IGFBP IGF-I kcal LBM LH NEB NEEE PYY RM T3 TEEE WEE American College of Sports Medicine Body mass index Body weight Energy availability Energy intake Fat-free mass Growth hormone Gonadotropin-releasing hormone Hypothalamic-pituitary-gonadal IGF-binding protein Insulin-like growth factor-I Kilocalories Lean body mass Luteinizing hormone Negative energy balance Non-exercise energy expenditure Peptide YY Resting metabolism Tri-iodothyronine Total energy expended during exercise Waking energy expenditure A. B. Loucks (*) Biological Sciences, Ohio Musculoskeletal and Neurological Institute, Ohio University, Athens, OH, USA e-mail: loucks@ohio.edu Introduction: The Female Athlete Triad This chapter summarizes the studies in our laboratory and others that identified low energy availability as the key factor causing the Female Athlete Triad and identifies four distinct origins of low energy availability among female athletes. In 2007, the American College of Sports Medicine (ACSM) published a revised position stand on the Female Athlete Triad [1], which replaced its earlier position stand on the same subject [2]. The revised position stand corrected the former misunderstanding of the Triad as a narrow syndrome consisting of disordered eating, amenorrhea, and osteoporosis by describing the Triad more broadly as the harmful effects of low energy availability on menstrual function and bone mineral density. The revised position stand emphasized that energy availability can be severely reduced by exercise energy expenditure alone without clinical eating disorders, disordered eating, or even dietary restriction. It also explained that low energy availability induces more menstrual disorders than amenorrhea and that these functional hypothalamic menstrual disorders must be carefully distinguished by differential diagnosis from other kinds of menstrual disorders not caused by low energy availability that are, therefore, unrelated to the Triad. The revised position stand also explained that bone mineral density in young athletes must be quantified in terms of Z-scores © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_11 171 172 instead of T-scores and that during adolescence low energy availability can cause Z-scores to decline as T-scores increase. Subsequently, treatment and return to play guidelines for the Triad were published in 2014 by the Female Athlete Triad Coalition [3, 4]. In 2014, an International Olympic Committee consensus statement introduced the term Relative Energy Deficiency in Sport to extend the concept of the Female Athlete Triad to include effects of energy deficiency beyond the reproductive and skeletal systems in men as well as women [5]. This chapter focuses on effects of low energy availability on reproductive function, specifically in women. Hypothetical Mechanisms of Functional Hypothalamic Menstrual Disorders in Exercising Women As in other fields of research, competing schools of thought developed to explain the high prevalence of menstrual disorders observed in exercising women. Of the several early mechanisms proposed, three were most widely held. Body Composition In 1974, body composition was offered as an explanation for the amenorrhea observed in anorexia nervosa patients [6]. This idea was a refinement of an earlier hypothesis about body weight accounting for the timing of menarche [7]. The body composition hypothesis held that menarche occurs in girls when the amount of energy stored in their bodies as fat rises to a critical 17% of their body weight, and that menstrual function is lost later when their body fat declines to less than a critical 22% of body weight [6]. The body composition hypothesis was the most widely publicized explanation for menstrual disorders in athletes in the lay community and the most widely embraced by the clinical community, even though it was the least widely accepted within the scientific community. The hypothesis was based entirely on correlations without any supporting experimental evidence A. B. Loucks [8]. Actually, observations of athletes did not consistently verify an association of menstrual status with body composition (e.g., Ref. [9]) and did not display the correct temporal relationship between changes in body composition and menstrual function (for reviews, see [10–13]). Rather, eumenorrheic and amenorrheic athletes were found to span a common range of body composition [14] leaner than that of eumenorrheic sedentary women. In addition, after the growth and sexual development of prepubertal animals had been blocked by dietary restriction, normal luteinizing hormone (LH) pulsatility resumed only a few hours after ad libitum feeding was permitted, before any change in body weight or composition could occur [15]. Moreover, when surgical reduction of the stomachs of severely obese women (body weight ~130 kg; body mass index [BMI] ~47) reduced the amount of food that they could eat, rapid weight loss and amenorrhea occurred while the patients were still obese (body weight ~97 kg; BMI ~35) [16]. Despite such criticisms, scientific interest in the body composition hypothesis was renewed with the discovery in 1994 of the adipocyte hormone leptin [17], with the observation of statistically significant correlations between leptin levels and body fatness in rodents and humans (e.g., Ref. [18]) and with the discovery of leptin receptors on hypothalamic neurons. Since then, an abundance of experimental evidence from rodents and human has demonstrated that a minimal level of leptin is permissive (i.e., necessary but not sufficient) for sexual development and function [19]. This permissive effect occurs indirectly via receptors on hypothalamic kisspeptin neurons that communicate with the hypothalamic gonadotropin-releasing hormone (GnRH) neurons that regulate LH pulsatility [20]. A 9-month double-blind, randomized, clinical trial administered pharmacological doses of leptin to women with functional hypothalamic amenorrhea whose BMI was in the range 18–25 kg/ m2 [21]. Prior to treatment, their leptin levels (mean ± SD = 4.6 ± 2.0 ng/ml) were within the lower portion of the range (7.4 ± 3.7 ng/ml) cited by the leptin assay manufacturer (Millipore Corp.) for women in this range of BMI [22]. Leptin levels comparable to those reported by the manufacturer 11 Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function have been found in other women with similar ranges of BMI [23–29]. The leptin dosages administered to the women with functional hypothalamic amenorrhea in this experiment raised their leptin levels more than tenfold (mean ± SD = 59 ± 37 ng/ml). Yet menstrual cycles occurred only intermittently, with the number of menstruating women fluctuating from month to month between 3 of 10 (30%) and 4 of 7 (57%). By contrast, nutritional counseling has restored spontaneous menstrual cycles in 75% of women with functional hypothalamic amenorrhea within 5 months [30]. Although leptin was originally thought to communicate information about fat stores, it was later found to vary profoundly in response to fasting, dietary restriction, refeeding after dietary restriction, and overfeeding before any changes in adiposity occurred [31–34]. This led to the hypothesis that leptin also signals information about dietary intake and specifically carbohydrate intake after leptin synthesis was found to be regulated by the tiny flux of glucose through the hexosamine biosynthesis pathway in both muscle and adipose tissue [35]. In eumenorrheic and amenorrheic athletes, leptin was found to differ not in its average concentration, but rather in the presence and absence, respectively, of a diurnal rhythm [23], and the diurnal rhythm was found to depend not on energy intake but rather on energy availability or more specifically on carbohydrate availability [27]. Thus, if leptin does participate in the functional regulation of the GnRH pulse generator in exercising women, it seems more likely to do so as a signal of low energy or carbohydrate availability than as a signal of low energy stores. Energy Availability In 1980, Warren was the first to suggest that menstrual function in dancers might be disrupted by an “energy drain” [36], but an empirically testable energy availability hypothesis was first clearly stated in terms of brain energy availability by Winterer, Cutler, and Loriaux in 1984 [37]. They hypothesized that failure to provide sufficient metabolic fuels to meet the energy requirements of the brain causes an alteration in brain 173 function that disrupts the GnRH pulse generator, although the mechanism of this alteration was unknown. At the organismal level, the energy availability hypothesis recognizes that mammals partition energy among several major metabolic activities, including cellular maintenance, immunity, thermoregulation, locomotion, growth, and reproduction [38] and that the expenditure of energy in one of these functions, such as locomotion, makes it unavailable for others, such as reproduction. Considerable observational data from biological field trials supports this idea and indicated that the dependence of reproductive function on energy availability operates principally in females (For reviews, see [38–42]. Experiments had induced anestrus in Syrian hamsters by food restriction, by the administration of pharmacological blockers of carbohydrate and fat metabolism, by insulin administration (which shunts metabolic fuels into storage), and by cold exposure (which consumes metabolic fuels in thermogenesis) [38]. Disruptions of reproductive function were independent of body size and composition. The energy availability hypothesis was also supported by endocrine observations of athletes. Amenorrheic athletes displayed low blood glucose levels during the feeding phase of the day [43], low insulin and high IGF binding protein-1 (IGFBP) during the fasting phase [43], loss of the leptin diurnal rhythm [23], high fasting acylated ghrelin [44], high peptide YY (PYY) [45], and low tri-iodothyronine (T3) levels in the morning [46, 47]. All of these abnormalities in metabolic substrates and hormones are signs of energy deficiency. T3 regulates basal metabolic rate, and low T3 occurs in numerous conditions, from fasting to cancer, in which dietary energy intake is insufficient to meet metabolic demands. In addition, eumenorrheic and amenorrheic athletes both displayed low insulin and high IGFBP-1 levels during the feeding phase of the day, as well as low leptin [23] and elevated growth hormone (GH) levels over 24 hours [43]. Indeed, eumenorrheic and amenorrheic athletes were found to be distinguished not by different 24-hour mean concentrations of leptin but rather by different amplitudes in the diurnal rhythm of leptin [23]. 174 Amenorrheic and eumenorrheic athletes reported similar stable body weights, despite dietary energy intakes similar to those of sedentary women [46, 48–52]. That is, they reported their dietary energy intakes to be much less than would be expected for an athlete’s level of physical activity. This apparent discrepancy between stable body weight and unexpectedly low dietary energy intake was controversial. Since energy intake and expenditure are very difficult to measure accurately, the apparent discrepancy might have been attributable to methodological errors. Some investigators attributed the apparent discrepancy between energy intake and expenditure in athletic women to underreporting of dietary intake [53, 54], because such underreporting is common in all populations [55], but underreporting did not account for the abnormalities in metabolic substrates and hormones observed in athletes. Furthermore, behavior modification and endocrine-mediated alterations of resting metabolic rate operate to stabilize body weight despite dietary energy excess and deficiency [56]. Exercise Stress The exercise stress hypothesis held that exercise disrupts the GnRH pulse generator by activating the hypothalamic–pituitary–adrenal axis. In order for the stress hypothesis to be meaningfully independent of the energy availability hypothesis, however, the adrenal axis must be activated independently of the energy cost of the exercise. Certainly, there are central and peripheral mechanisms by which the adrenal axis can disrupt the ovarian axis [57], and prolonged aerobic exercise without glucose supplementation does activate the adrenal axis. Selye first induced anestrus and ovarian atrophy in rats by abruptly forcing them to run strenuously for prolonged periods [58]. Later, others also induced anestrus by forced swimming [59, 60], by forced running [61], and by requiring animals to run farther and farther for smaller and smaller food rewards [62, 63]. The elevated cortisol levels induced in such experiments were interpreted as signs of stress, and the resulting disruptions of the hypothalamic-­ A. B. Loucks pituitary-­gonadal (HPG) axis were widely interpreted as evidence that “exercise stress” has a counter-regulatory influence on the female reproductive system. Amenorrheic athletes also display mildly elevated cortisol levels [43, 48, 64–66]. This observation was the basis for attributing their amenorrhea to stress. Mild hypercortisolism is also associated with amenorrhea in patients with functional hypothalamic amenorrhea [67] and anorexia nervosa [68]. This interpretation overlooked the glucoregulatory functions of cortisol, which inhibit skeletal muscle glucose uptake and promote skeletal muscle proteolysis for hepatic gluconeogenesis in response to low blood glucose levels [69]. Thus, it was possible that the mild hypercortisolism observed in amenorrheic athletes might have reflected a chronic energy deficiency rather than exercise stress. At the time, it was not known whether the adrenal cortical axis mechanisms that disrupt the HPG axis in forced exercise experiments on animals also operate in voluntarily exercising women. Indeed, up to that time, all animal experiments investigating the influence of the “activity stress paradigm” on reproductive function had confounded the stress of exercise with the stress of the method used to force animals to exercise. These experiments had also been confounded by the energy cost of the exercise performed, and glucose supplementation during exercise was found to blunt the usual rise in cortisol in both rats [70] and men [71]. As a result, in 1990 the literature on stress contained only ambiguous evidence that the stress of exercise disrupts the HPG axis in either animals or humans. Prospective Clinical Experiments xperiments Confounding Exercise E Stress and Energy Availability Several investigators attempted to induce menstrual disorders through chronic exercise training, but most [72–75] applied only a moderate volume of exercise, or the volume of exercise was increased gradually over several months, and 11 Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function diet was uncontrolled or unquantified. One study [75] selected physically trained subjects who appeared to have been luteally suppressed before the study even began [76]. Only one experiment had successfully induced menstrual disorders in regularly menstruating women [77]. Modeled on Selye’s early animal experiments [58], this single successful experiment imposed a high volume of aerobic exercise abruptly, thereby suppressing follicular development, the LH surge, and luteal function in a large proportion of the subjects in the first month and in an even larger proportion in the second. Both proportions were greater in a subgroup fed a controlled weight loss diet than in another subgroup fed for weight maintenance, but even the weight maintenance subgroup may have been underfed, since behavior modification and endocrine-­ mediated alterations of resting metabolic rate operate to stabilize body weight despite dietary energy excess and deficiency [56]. Such experiments, in which outcome variables are properties of the menstrual cycle, require sustained observations over a period of several weeks. Such prolonged experimental protocols suffer from practical problems with subject retention and compliance with experimental treatments. To avoid these difficulties, shortterm experimental protocols were developed in which LH pulsatility was chosen as the outcome variable, because ovarian function is critically dependent on LH pulsatility. Of course, shortterm effects on LH pulsatility are not proof of chronic effects on ovarian function, but hypotheses about mechanisms regulating LH pulsatility could be tested in highly controlled short-term experiments, and then chronic effects could be confirmed in prolonged experiments later. One such short-term experimental protocol found that a combination of increased exercise and dietary restriction disrupts LH pulsatility during the early follicular phase [78]. LH pulse frequency during 12 waking hours was lower in four habitually physically active women when their exercise training regimen was increased during a few days of dietary restriction than during dietary supplementation. However, this experiment did not determine whether LH pulse 175 frequency could be suppressed by exercise without dietary restriction or whether the stress of exercise had a suppressive effect on LH pulsatility beyond the impact of the energy cost of exercise on energy availability. Experiments Distinguishing the Independent Effects of Exercise Stress and Energy Availability For several years, we focused our efforts on a series of studies that we called the “Excalibur” experiments that were designed to determine the independent effects of exercise stress and energy availability on the HPG axis [28, 29, 79–83]. For these experiments, we defined energy availability operationally as dietary energy intake minus exercise energy expenditure. Conceptually, this corresponds to the amount of dietary energy remaining after exercise training for all other physiological functions. Although not the actual physiological quantity hypothetically affecting the HPG axis at the cellular level, our operational definition in behavioral terms had the advantage of being readily measurable and controllable. We controlled the dietary energy intake of our subjects by feeding them diets of known amount and composition as their only food during the experiments. We also required them to exercise under supervision in our laboratory on a treadmill while we measured and controlled their energy expenditure until they had expended a predetermined amount of energy. In the absence of any empirically operational definition of stress [84], we defined exercise stress independently as everything associated with exercise except its energy cost. Through careful subject selection, we took steps to minimize the influence of potentially confounding factors. Healthy, regularly menstruating, habitually sedentary, nonobese, non-­ smoking women 18–34 years of age at least 5 years past menarche, with no recent history of dieting, weight loss, or aerobic training were recruited. Before being admitted to the study, these volunteers underwent an extensive screening procedure, including written medical, men- A. B. Loucks 176 strual, dietary, and athletic histories, a physical examination, a 12-lead resting electrocardiogram, a 7-day prospective dietary record, determination of body composition by hydrostatic weighing or whole body air-displacement plethysmography, and a treadmill test to determine their aerobic capacity. Volunteers were admitted into experiments only if they presented no current use of medications including oral contraceptives and no history of heart, liver, or renal disease, diabetes, and menstrual or thyroid disorders. They must also have had documented prospective records of menstrual cycles 26–32 days in length for at least the previous 3 months. They were required to be 18–30% body fat, with habitual energy intakes between 35 and 55 kcal/kg lean body mass (LBM)/day based on their 7-day diet records, with maximal aerobic capacities less than 42 ml O2/kg body weight (BW)/min, and they must have been performing less than 60 minutes of habitual aerobic activity per week for the previous 3 months. The narrow range of our subjects’ menstrual cycle lengths implied that we restricted our subject pool to the central 60% of menstrual cycle lengths in the population and that from this pool we chose women whose menstrual cycle lengths were in the least variable 20% of the population [85]. Thus, if anything, our subjects’ reproductive systems were robust against disturbance by commonly occurring environmental and behavioral influences. We could be confident, therefore, that if our treatments disrupted the reproductive systems of these women, they would disrupt the reproductive systems of other women, too. We could also be confident that our subjects’ metabolism had not been disturbed by any confounding medical conditions or dietary or exercise habits before our treatments were applied. Excalibur I Excalibur I [79] was designed to investigate whether exercise stress had any suppressive effect on T3 levels independent of the impact of the energy cost of exercise on energy availability. We were interested in T3 because it regulates the rate of energy expenditure at rest and because it was known to be suppressed in amenorrheic athletes. We reasoned that if the energy cost of exercise necessitates such major metabolic adjustments as the suppression of reproductive function, then these metabolic adjustments might be mediated in part by suppressing T3. Over the course of the Excalibur experiments, our insight into how to correctly quantify energy availability (EA) for subjects of various body sizes gradually matured. At the time of Excalibur I, we normalized energy intake (EI) and exercise energy expenditure to body weight (BW). We also measured exercise energy expenditure as the total energy expended during exercise (TEEE), as measured by an ergometer. EA = ( EI − TEEE ) / BW In Excalibur I, we found that severely low energy availability (8 kilocalories per kilogram of body weight per day, kcal/kgBW/day) suppressed T3 levels by 15%, while exercise stress had no effect on T3. T3 levels were suppressed similarly regardless of whether energy availability was reduced by dietary energy restriction or by exercise energy expenditure. Furthermore, the suppression of T3 in exercising women was prevented by supplementing their diet in compensation for the energy cost of their exercise. These findings were consistent with the energy availability hypothesis and inconsistent with the exercise stress hypothesis. Excalibur II Excalibur II [80] was designed to reveal whether T3 levels in exercising women vary in linear proportion to energy availability or are suppressed abruptly at a particular threshold of energy ­availability. By this time we had realized that very little energy expenditure occurs in body fat. Accordingly, we changed our normalization of energy intake and expenditure to lean body mass (LBM) which would exclude body fat. EA = ( EI − TEEE ) / LBM We administered various levels of energy availability to exercising women and found that the suppression of T3 by low energy availability occurred 11 Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function abruptly at a threshold of energy availability near 25 kcal/kgLBM/day. For our women of average body size (59 kg) and composition (24.5% body fat), that threshold was about 1000 kcal/day. Excalibur III Normal ovarian function depends not on some stable concentration of LH but rather on the occurrence of pulsatile surges of LH concentrations in the blood at regular intervals. These pulses correspond to regular secretory bursts of LH from the pituitary gland in response to similar secretory bursts of GnRH from the hypothalamus. The frequency (at intervals of 70–180 minutes) and amplitude of these pulses vary around the menstrual cycle. In sedentary women in the early follicular phase, the pulsatile pattern is characterized as high frequency and low amplitude. In regularly menstruating athletes, the pulses occur less often and are larger in amplitude but still at regular intervals. In amenorrheic athletes, LH pulses occur even less often and irregularly [48]. Therefore, in Excalibur III [81, 82], we investigated whether exercise has any suppressive effect on LH pulsatility beyond the impact of its energy cost on energy availability. The design of Excalibur III is illustrated in Fig. 11.1. For 4 days in the mid-follicular phase of two menstrual cycles, we controlled the energy availability of two groups of women. During one cycle, we administered a balanced energy availability of 45 kcal/kgLBM/day, and during the other cycle, we administered a low energy availability of 10 kcal/kgLBM/day. One group of subjects performed no exercise during the two treatment periods. A second group performed the same large volume of high-intensity exercise that we had utilized in Excalibur I (30 kcal/kgLBM/day at 70% VO2max; maximal aerobic capacity]). We imposed balanced and low energy availabilities on the non-exercising group by feeding them 45 and 10 kcal/kgLBM/day, respectively. We imposed the same balanced and low energy availabilities on the group performing 30 kcal/ kgLBM/day of exercise by feeding them 75 and 40 kcal/kgLBM/day, respectively. Between Excalibur II and III, we had had another insight into the proper quantification of energy availability. Prior to Excalibur III [81, 82], 90 90 45 45 B 0 kcal . kg–1 FFM Fig. 11.1 Experimental design of Excalibur III. Dietary energy intake (I) and exercise energy expenditure (E) were controlled to achieve balanced (B = 45 kcal/kgLBM/ day) and deprived (D = 10 kcal/kgLBM/ day) energy availability (A = I-E) treatments. Deprived energy availability was achieved by dietary restriction alone in sedentary women (S) and by exercise energy expenditure alone in exercising women (X) (1 kcal = 4.18 kJ). (Reproduced with permission from [82]) 177 0 -45 I E I A E A -45 90 90 45 45 D 0 0 -45 -45 S X A. B. Loucks 178 we had calculated energy availability by subtracting total energy expenditure during exercise (TEEE) from dietary energy intake. While we were designing Excalibur III, however, we recognized that if our exercising subjects had not been exercising, their other routine activities during the same hours would have resulted in some non-­exercise energy expenditure (NEEE). Therefore, the actual energy expenditure due to exercise itself (EEE) was less than the total energy expenditure measured during exercise (EEE = TEEE – NEEE). This adjustment would be especially important for Excalibur III, in which some subjects exercised and others did not. So, in Excalibur III and our later experiments, we changed again the way we calculated energy availability by subtracting from dietary energy intake only the portion of total energy expenditure during exercise that was directly attributable to the exercise itself. during exercise that was specifically attributable to the exercise. In retrospect, our subjects’ energy expenditure in routine activities on a non-­ exercising day during the same 3 hours when they exercised in Excalibur II had amounted to 5 kcal/kgLBM/day, and our calculations of energy availability had been underestimated by the same amount. At the end of each of the 4-day treatments in Excalibur III, we admitted the women to a general clinical research center and drew blood samples from them at 10-minute intervals for 24 hours. Later, we measured the amount of LH in each sample and used a special statistical computer program to detect and to calculate the frequency and amplitude of their LH pulses. We determined the effects of energy availability on these frequencies and amplitudes by contrasting data taken while performing the same exercise at different energy availabilities, and we determined the independent effect of exercise stress by conEA = ( EI − EEE ) / LBM trasting groups exercising differently at the same We achieved this by using an activity monitor energy availabilities. to measure our subjects’ energy expenditure in We found that low energy availability reduced their normal daily activities during the same LH pulse frequency and increased LH pulse hours of the day when they would be exercising amplitude, while exercise stress had no suppresin our experiment. We then subtracted this non-­ sive effect on LH pulsatility beyond the impact of exercise energy expenditure in routine activities the energy cost of exercise on energy availabilfrom their total energy expenditure during exer- ity (Fig. 11.2). LH pulsatility was disrupted by cise to obtain the amount of energy expenditure extreme energy restriction alone and by extreme 0 20 -1 15 Pulses . day–1 Fig. 11.2 Effects of low energy availability on LH pulsatility in Excalibur III. Left: Luteinizing hormone (LH) pulse frequency in sedentary (S) and exercising (X) women with the same balanced energy availability (45 kcal/kgLBM/day). Right: Reduction in LH pulse frequency caused by low energy availability (10 kcal/ kgLBM/day) in sedentary (S) and exercising (X) women. ∗ p < 0.01. (Adapted from [81, 82]) -2 * -3 10 -4 -5 5 -6 0 S X -7 * * S X 11 Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function exercise energy expenditure alone. Dietary supplementation prevented the suppression of LH pulsatility by exercise energy expenditure. Others have shown that short-term fasting also reduces LH pulse frequency in sedentary women during the early follicular phase [86, 87] and that in lean women, ovarian function is also impaired during the ensuing menstrual cycle [87]. In Excalibur III, low energy availability also suppressed plasma glucose, insulin, insulin-like growth factor-I (IGF-I), leptin, and T3 while raising growth hormone (GH) and cortisol levels. All these effects are reminiscent of abnormalities observed in amenorrheic athletes [43, 46–48, 64–66]. This contradiction of the exercise stress hypothesis has been confirmed by more prolonged experiments on animals. Amenorrhea was induced in monkeys by training them to run voluntarily on a motorized treadmill for longer and longer periods, while their food intake remained constant [88]. Then their menstrual cycles were restored by supplementing their diets without any moderation of their exercise regimen [89]. The exercise stress hypothesis was also contradicted in a novel animal model of the entire Female Athlete Triad [90]. In this modified activity stress paradigm, rats were habituated to voluntary wheel running for 90 days and then randomized to control and restricted diets for the next 90 days. Although both groups ran similar distances and expended similar amounts of energy in exercise, estradiol was suppressed, estrous cycling ceased, ovaries were atrophied, and the bone mineral content of the femur and tibia were reduced only in the underfed rats. The suppression of LH pulse frequency by low energy availability in Excalibur III was actually smaller in exercising women than in non-­ exercising women with the same low energy availability [82]. This result was unexpected, and it suggested that LH pulsatility might actually depend on a more specific metabolic factor that is easily confused with energy availability, but which is less compromised by exercise energy expenditure than by dietary energy restriction. Research in other mammals suggests that GnRH neuron activity and LH pulsatility are 179 actually regulated by brain glucose availability [38, 41]. The adult female human brain oxidizes about 80 g of glucose each day at a continuous rate. This must be provided daily by dietary carbohydrate, because the brain’s rate of energy expenditure can deplete liver glycogen stores in less than a day [91]. To that end, moderate exercise oxidizes as much glucose in an hour. In the non-exercising women in Excalibur III, low energy availability due to dietary energy restriction reduced carbohydrate intake by 77%. This reduction in carbohydrate intake was similar to the 73% increase in carbohydrate oxidation revealed by respiratory gas analysis in the exercising women during the balanced energy availability treatment. By contrast, carbohydrate oxidation increased only 49% in the exercising women under low energy availability conditions. This alteration in fuel selection conserved almost 70% of the brain’s daily glucose requirement. Thus, exercise may compromise brain glucose availability less than dietary energy restriction, and this may account for the smaller disruption of LH pulsatility that we observed in exercising women than in dietary-restricted women. Thus, LH pulsatility may depend specifically on carbohydrate availability rather than energy availability in women, just as it does in other mammals. Excalibur IV Excalibur IV [83] was designed to reveal whether refeeding reverses the suppression of LH pulsatility in women as quickly as it does in other mammalian species. In food-restricted female rats [15, 92] and ewes [93], and in fasted heifers [94] and male rhesus monkeys [95], a single ad libitum meal stimulates LH pulses within 2 hours. Such observations have been interpreted to imply that the physiological signals produced by a single large meal are sufficient to activate the hypothalamic GnRH neurons that control LH pulsatility [96]. We suspected that the restoration of LH pulsatility by refeeding might be considerably slower in energetically disrupted women than in other mammals, because the human brain requires so much more energy than does the brain of any other mammal. The brain competes against all 180 other tissues of the body for energy, and the adult human brain requires 20% of basal metabolic energy, compared to only 2% for most species and 8% for nonhuman primates [97]. Therefore, we suspected that a single meal might not provide enough energy to activate GnRH neurons in energetically disrupted women. To stringently test this hypothesis, we assayed LH in blood samples drawn from women at 10 minute intervals for 48 hours during the mid-­ follicular phase, first during 24 hours on the fifth day of low energy availability treatments and then during 24 hours of aggressive refeeding. A combination of moderate dietary energy restriction (25 kcal/kgLBM/day) and moderate exercise energy expenditure (15 kcal/kgLBM/day) was administered to impose a low energy availability of 10 kcal/kgLBM/day. The aggressive refeeding regimen was comprised of 15 meals providing a total of 85 kcal/kgLBM/day. Combined with the same exercise treatment, the energy availability during the 24 hours of aggressive refeeding was 70 kcal/kgLBM/day. Compared to measurements of LH pulsatility in 18 other women studied previously in our laboratory under balanced energy availability conditions and at the same phase of the menstrual cycle, low energy availability suppressed LH pulsatility unambiguously in five of the eight subjects treated in this experiment. Their LH pulse frequency was reduced 57% to 8.2 ± 1.5 pulses/24 hours, well below the 5th percentile of LH pulse frequencies in energy balanced women (14.6 pulses/24 hours), while their LH pulse amplitude was increased 94% to 3.1 ± 0.3 IU/L, well above the 95th percentile of LH pulse amplitudes in energy balanced women (2.5 IU/L). Amongst these women, aggressive refeeding raised LH pulse frequency by only 2.4 ± 1.0 pulses/24 hours, still far below the 5th percentile of LH pulse frequency in energy balanced women. Meanwhile, the unambiguously elevated LH pulse amplitude was completely unaffected (Δ = 0.0 ± 0.4 IU/L) by aggressive refeeding. Results were similar when all eight subjects were included in the analysis. Aggressive refeeding pushed the group as a whole to, but not past, the 5th and 95th percentiles of LH pulse A. B. Loucks frequency and amplitude, respectively. Thus, as we had suspected, 24 hours of a refeeding protocol much more aggressive than the ad libitum refeeding protocols commonly employed in animal experiments had very little restorative effect on LH pulsatility in our energetically suppressed women. Excalibur V In an experimental protocol similar to that of Excalibur II, Excalibur V determined the dose-­ response effects of low energy availability on LH pulsatility in habitually sedentary, regularly menstruating young women [28]. To do this, we administered balanced and one of three low energy availabilities (45 and either 10, 20, or 30 kcal/kgLBM/day) to healthy, habitually sedentary, regularly menstruating women for 5 days. The design is illustrated in Fig. 11.3. We found that LH pulsatility was disrupted within 5 days below a threshold of energy availability at ~30 kcal/kgLBM/day (Fig. 11.4). This was, in fact, the same actual energy availability that we had reported as 25 kcal/kgLBM/day in Excalibur II [80], because we had underestimated energy availability by 5 kcal/kgLBM/day in Excalibur II, as described in the discussion of Excalibur III above. The disruption of LH pulsatility below 30 kcal/kgLBM/day in Excalibur V was consistent with many observational studies of amenorrheic runners, all of which indicated energy availabilities less than 30 kcal/kgLBM/day [98]. It was also consistent with the only prospective study of the refeeding of amenorrheic athletes, in which menstrual cycles had been restored in runners by increasing their energy availability from 25 to 31 kcal/kgLBM/day [99]. Energy availabilities below 30 kcal/kgLBM/day have also been reported in eumenorrheic athletes [98], 80% of whom display subclinical ovarian disorders in which the suppression of progesterone may also impair fertility [100]. In the same experiment, we also determined the dose-response effects of low energy availability on several metabolic substrates and hormones. Down to an energy availability of 30 kcal/kgLBM/day, the responses of insulin, 11 Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function I) 60 Energy Intake and Expenditure (kcal/kgLBM/day) 181 50 d olle tar Die y erg n yE ( ake Int ntr Co 40 30 20 10 0 Controlled Exercise Energy Expenditure (X) 0 5 10 15 20 25 30 35 40 45 50 Energy Availability (kcal/kgLBM/day) Fig. 11.3 Experimental design of Excalibur V. Women were assigned to contrasting energy availability treatments of 45 and 10, 45 and 20, and 45 and 30 kcal/ kgLBM/day. All subjects performed a controlled exercise energy expenditure of 15 kcal/kgLBM/day in aero- 75 Percent (%) 50 A/3 25 0 -25 F -50 -75 0 10 20 30 40 50 Energy Availability (kcal/kgLBM/day) Fig. 11.4 Incremental effects of energy availability on LH pulse amplitude (A/3) and LH pulse frequency (F) in Excalibur V. Effects are expressed relative to values at 45 kcal/kgLBM/day. Effects on LH pulse amplitude have been divided by three for graphical symmetry. As energy availability declines from energy balance at approximately 45 kcal/kgLBM/day, effects begin at a threshold at approximately 30 kcal/kgLBM/day and become more extreme as energy availability is further reduced below 20 kcal/kgLBM/day. (Reproduced with permission from [28], Copyright 2003, The Endocrine Society) cortisol, insulin-like growth factor (IGF)-I/IGFbinding protein (IGFBP)-1, IGF-I/IGFBP-3, leptin, and T3 maintained plasma glucose lev- bic exercise at 70% VO2 max under supervision, while their dietary energy intake was controlled to achieve the intended energy availabilities. (Reproduced with permission from [28], Copyright 2003, The Endocrine Society) els to within 3% of normal values. Below that threshold, however, plasma glucose levels fell despite further increases in the responses of the metabolic hormones, and effects on LH pulsatility appeared. Excalibur V also revealed the dose-response effects of low energy availability on biochemical markers of bone turnover [101]. Urinary concentrations of N-telopeptide of type I collagen, a marker of the rate of whole body bone resorption, rose as estradiol concentrations declined, when energy availability was lowered to 10 ­kcal/ kgLBM/d. By comparison, markers of bone formation declined at higher energy availabilities. Concentrations of serum carboxy-terminal propeptide of type I procollagen, a marker of bone type I collagen synthesis, and insulin declined linearly with energy availability. By contrast, concentrations of osteocalcin, a marker of bone mineralization, declined abruptly below 30 kcal/ kgLBM/day together with IGF-I and T3, which modulates the hepatic synthesis of IGF-I in response to GH stimulation. Such uncoupling of bone turnover, with increased resorption and reduced formation, can lead to irreversible reductions in bone mineral density [102]. 182 Excalibur VI The prevalence of amenorrhea has been reported to decline from 67% in marathon runners younger than 15 years of gynecological age to only 9% in those who were older [103]. Meanwhile, in the general population, the incidence of menstrual disorders declines during the decade after menarche as fertility increases [104]. Excalibur VI investigated whether these two observations might both be explained by a declining sensitivity of LH pulsatility to low energy availability as the energy cost of growth decreases [29]. Calcium balance, which is an index of growth, does not decline to zero until 14 years of gynecological age [105]. In Excalibur VI, contrasting balanced and low energy availabilities (45 and 10 kcal/kgFFM/day) were administered to healthy, habitually sedentary, regularly menstruating, older adolescent women (5–8 years of gynecological age, ~20 years of calendar age) and young adult women (14–18 years of gynecological age, ~29 years of calendar age) for 5 days. Low energy availability suppressed LH pulsatility in the adolescents but not in the adults, even though metabolic and endocrine signals of energy deficiency (i.e., plasma glucose, β-hydroxybutyrate, insulin, cortisol, T3, leptin, IGF-1, and GH) were altered as much or more in the adults as in the adolescents [29]. This insensitivity of LH pulsatility to energy deficiency in adult women was subsequently confirmed by a corresponding insensitivity of ovarian function to energy deficiency [106]. In that experiment, the energy availability of women 25–40 years of age was reduced to ~25 kcal/ kgFFM/day for 4 months by a combination of dietary restriction (~600 kcal/day) and exercise (~200 kcal/day). This subthreshold energy deficiency reduced the body fatness of these reproductively mature women from 32% to 27% but caused no more than a mild suppression of luteal function. An adult reproductive system that is more robust against insults of energy deficiency may be explained by a greater availability of glucose to the brain in adults than in adolescents at the same energy availabilities. This might occur if peripheral tissues in full-grown adults do not compete as aggressively against the brain for A. B. Loucks available energy or carbohydrate. Alternatively, the sensitivity of sensors in the central nervous system to signals of energy deficiency may decline during adolescence. These possibilities remain to be investigated. ther Efforts to Manipulate Energy O Availability in Habitually Sedentary Women A recent study by Lieberman et al. [107] investigated the effects of energy availability on menstrual function by reanalyzing data collected in an earlier experiment that had attempted to administer controlled negative energy balance (NEB) treatments of −15%, −30%, and −60% to separate groups of habitually sedentary regularly menstruating women for 3 months [108]. Thirty-­ five women with 5–15 years of gynecological age and ovulatory cycles as long as 35 days were studied, even though cycles of 36 days were to be classified as a clinical menstrual disturbance (oligomenorrhea) and the average within-person annual standard deviation of cycle length at the subjects’ age is 4 days [85]. In practice, NEB turned out to be less negative and more widely dispersed than intended (mean ± 2SD, −8 ± 10%, −22 ± 21%, and −42 ± 9%). Moreover, metabolic hormone indicators of energy deficiency did not display dose-­ response effects of group differences in NEB. Assuming the underlying diet and exercise data were correct, Lieberman et al. calculated energy availability values in each menstrual cycle and found a continuum of energy availability treatments from 18 to 51 kcal/kgFFM/day. Unfortunately, Lieberman et al. did not report the effects on metabolic hormones. They found no dose-response effects of energy availability on ovarian steroids. Altogether, they found that 36% of 105 menstrual cycles across the range of energy availability displayed menstrual disturbances and 85% of these were subclinical (luteal phase deficiency and anovulation). Collectively, only one menstrual cycle was missed. Lieberman et al. concluded that their results “do not support that a threshold energy availability exists below which menstrual disturbances are induced,” thereby appearing to confirm the 11 Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function Female Athlete Triad as a continuum of interrelated disorders. However, given the 10–15% incidence of oligomenorrhea and the 15–65% incidence of subclinical menstrual disturbances in free-living women of the same gynecological age [109, 110], the observations of Lieberman et al. are better interpreted as what would be expected without any intervention. Moreover, without a crossover design and without dose-­ response effects of energy availability on any physiological indicator of energy deficiency, Lieberman et al. simply lack evidence that the disturbances they observed were caused by the treatments administered. Reversal of Amenorrhea in Amenorrheic Athletes Cialdella-Kam et al. [111] administered a carbohydrate-­ protein dietary supplement of 360 kcal/day to athletes with clinical menstrual disorders (7 amenorrheic and 1 oligomenorrheic). After 6 months, the eight athletes had resumed menses with seven of them resuming ovulation. However, it should be noted that the investigators calculated EA without subtracting non-exercise energy expenditure NEEE. Therefore, as they acknowledged in another paper [112], their pre and post EA values probably underestimated actual EA values by 1–2 kcal/kgFFM/day. Prior to this study, there had been pilot studies published of amenorrheic athletes who increased caloric intake for several months, and changes in menstrual status were observed [99, 113]. Pre and post EA values depended, of course, on the definition of exercise. When exercise was defined as activity when energy expenditure was greater than 4.0 METS, the dietary supplement increased energy availability from 37 to 45 kcal/ kgFFM/day (p = 0.10). However, when exercise was defined more broadly to include all planned exercise plus bicycle commuting and all walking, energy availabilities were lower with the dietary supplement increasing them from 28 when amenorrheic to 39 kcal/kgFFM/day after restoration of menses (p = 0.09) [112]. More prospective research is needed to determine successful behavioral strategies that amenorrheic athletes with low energy availability can 183 use to resume menstrual status. For example, research on appetite suppression by exercise and dietary restriction suggests that it may be important for athletes to consume planned amounts of energy at planned times, by discipline instead of appetite [114]. onclusions About the Hypothetical C Mechanisms of Functional Hypothalamic Amenorrhea in Female Athletes We are unaware of any experiments that have determined the independent effect of body composition on the HPG axis. From the available experimental data, however, it would appear to be more likely that a lean body composition and disruption of the HPG axis are both effects of low energy availability than that a lean body composition disrupts the HPG axis. Our shortterm experiments on women have demonstrated that exercise stress has no suppressive effect on LH pulsatility beyond the impact of the energy cost of the exercise on energy availability. These short-term 4–5-day experiments investigating the independent effects of exercise stress and low energy availability on LH pulsatility predicted and, as we expected, were later confirmed by long-term experiments investigating the ­independent effects of exercise stress and low energy availability on estrus and menstrual cycles. Prospective controlled experiments on both humans and animal models have demonstrated that the factor disrupting the HPG axis in physically active women is low energy availability. These experiments suggest that women may be able to prevent or to reverse menstrual disorders by dietary reform alone without moderating their exercise regimen. As long as dietary energy intake is managed to keep energy availability above 30 kcal/kgLBM/day, there may be no need to interfere with endurance, strength, and skill training. Finally, the susceptibility of women to the disruption of reproductive function by energy deficiency appears to be substantially greater in those younger than 15 years of gynecological age. 184 auses of Low Energy Availability C in Female Athletes Effective treatment of low energy availability in athletes requires that the origin of the low energy availability be identified. Low energy availability behaviors appear to derive from four different origins [1, 114]. Some athletes intentionally reduce energy availability in a rational, but misguided, pursuit of the body size, body composition, and mix of metabolic fuel stores that are thought to optimize performance in their particular sport. Complex objectives may include reducing fat mass while increasing muscle mass and maximizing glycogen stores. For such athletes who reduce energy availability excessively, nutrition education and guidance regarding appropriate, individualized intermediate and ultimate goals, schedules, and methods may be sufficient to modify their diet and exercise behavior. In other athletes, low energy availability originates in an eating disorder. Eating disorders are clinical mental illnesses that are often accompanied by other mental illnesses [115, 116]. Therefore, eating disorders require psychiatric treatment, often inpatient treatment, as well as nutritional counseling. Because the mortality of eating disorders is so high, sports organizations need to develop institutional methods for distinguishing undernourished athletes with eating disorders from those who do not have eating disorders. This distinction may not be obvious, since undernourished athletes who are only trying to optimize performance may practice many of the same disordered eating behaviors (e.g., skipping meals, vomiting, using laxatives, etc.) as athletes with eating disorders. Athletes with eating disorders are distinctive in their resistance to the efforts of coaches, trainers, nutritionists, and physicians to modify their behavior. The third origin of low energy availability in athletes is the suppression of appetite by prolonged exercise. This effect is compounded by the appetite-suppressing effect of diets containing high percentages of carbohydrates, which are commonly recommended to athletes in endurance sports. Even though many studies on this subject have been published over the past 20 years [114, A. B. Loucks 117], appetite remains a largely neglected topic in the field of sports nutrition. Indeed, the word “appetite” appears only twice, in the recently revised joint position stand of the American Dietetic Association, the Dietitians of Canada, and the American College of Sports Medicine on nutrition and athletic performance [118]. Briefly, food deprivation increases hunger, but the same energy deficit produced by exercise energy expenditure does not [119]. The appetite-­ suppressing effect of prolonged exercise has been demonstrated in controlled experiments with protocols ranging from a few hours to 12 weeks [114]. The effect is mediated by the orexigenic hormone ghrelin, which induces us to begin eating, and by several anorexigenic hormones (including peptide YY, glucagon-like peptide 1, and pancreatic polypeptide) that induce us to stop eating. Exercise does not stimulate an increase in ghrelin concentrations but does stimulate increases in the concentrations of anorexigenic hormones (see associated Chaps. 12 and 30). As a result, “there is no strong biological imperative to match energy intake to activity-induced energy expenditure” [120]. Meanwhile, the appetite-suppressing effect of diets containing high percentages of c­ arbohydrates has been demonstrated in experimental protocols ranging from a week [121] to a month [122, 123]. As the percentage of carbohydrates in the diet was reduced, ad libitum energy intake spontaneously increased. As a result, the actual amount of carbohydrate consumed was preserved even though the percentage of carbohydrates in the diet decreased from 67% to 55%. The mechanism of this effect has not yet been identified but may involve the greater bulk and fiber content of carbohydraterich foods. Importantly, the large effects of these two factors are additive [121] so that together they can reduce energy availability below 30 kcal/kgFFM/ day in endurance athletes. To avoid inadvertent low energy availability, therefore, athletes in endurance sports need to be trained to eat by discipline (i.e., planned amounts of selected foods at scheduled times) instead of appetite. The fourth apparent origin of low energy availability among female athletes is that young 11 Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function women under-eat for social reasons unrelated to sport. Around the world, about twice as many young women as young men at every decile of body mass index perceive themselves to be overweight, and the numbers actively trying to lose weight are even more disproportionate [124]. Alarmingly, the disproportion even increases as BMI declines, so that almost 9 times as many lean women as lean men are actively trying to lose weight! Indeed, more young female athletes report improvement of appearance than improvement of performance as a reason for dieting [125]. As a result, social issues unrelated to sport may need to be addressed to persuade female athletes to eat by discipline beyond their appetites. ources of Error in the Estimation S and Control of EA In publications of the Excalibur experiments, the portion of body composition apart from fat mass is termed LBM. It is better termed fat-free mass (FFM). Then, as currently understood, energy availability (EA) is quantified by measuring dietary energy intake (EI), exercise energy expenditure (EEE), and fat-free mass (FFM). EA is then calculated as: 185 EA = ( EI − EEE ) / FFM A common source of error (by us in Excalibur I and II and by others) in studies of EA in athletes has derived from the misunderstanding of EEE as the total energy expenditure that would be measured by an ergometer during exercise. This misunderstanding has led to underestimations of EA, misinterpretations of data, and unwarranted criticisms of the concept. As described in the discussion of Excalibur III above, EEE is defined as the extra energy expended beyond the energy that would have been expended if no exercise had been performed (see Fig. 11.5). Defining EEE in this way enables EA to be fairly compared between different groups of subjects who do and do not exercise and between repeated observations of the same subjects when they do and do not exercise. Because energy expenditure varies with routine activities during the day, to calculate EA consistently with the Excalibur experiments, non-­ exercise energy expenditure (NEEE) must be measured on another non-exercising day during the same waking hours when exercise is performed. Then EEE is calculated as the difference between total energy expenditure during exercise (TEEE) and NEEE on the other day: EE EEE Non-Exercise Waking Activity Resting Metabolism = 30 kcal/kgFFM/day 00 03 06 09 12 Fig. 11.5 Calculation of exercise energy expenditure (EEE). (A. Top) EEE is the amount of energy that a woman expends because she is an athlete and does not include the energy she expends in resting metabolism and other waking activities. (B. Middle) Ergometers measure total energy expenditure during exercise (TEEE), which overestimates EEE by ~2 kcal/kgFFM/d per hour of exer- 15 18 Hrs 21 24 cise. For high-intensity exercise of short duration, the resulting error in calculating energy availability as EA = (EI – TEEE)/FFM is negligibly small for clinical purposes. (C. Bottom) For low-intensity exercise of long duration, however, the error in EA = (EI – TEEE)/FFM is very large and will lead to unwarranted changes in diet and exercise behavior. (Adapted from [126]) A. B. Loucks 186 EEE = TEEE − NEEE In an example described in a previous review [126], the resting metabolism (RM) of an athlete in energy balance on a non-exercising day is assumed to be 2/3 of her EI. For EI = 2100 kcal/ day (8.8 MJ/day), RM = 1400 kcal/day (5.8 MJ/ day) or 58 kcal/hour (244 kJ/h). If she sleeps 8 hours, her routine activities in waking energy expenditure (WEE) would expend the rest of her EI. Ignoring for simplicity other sources of diurnal variation in energy expenditure, her average rate of WEE would be 700 kcal/16 hours = 44 kcal/h (182 kJ/h). If her fat-free mass (FFM) is 45 kg, then her rate of non-exercise energy expenditure (NEEE) during exercise would be: NEEE = ( RM + WEE ) / FFM = ( 58 + 44 ) / 45 = 2.3 kcal / kgFFM / h ( 9.5 kJ / h ) If the athlete’s total energy expenditure during a 40-minute run is TEEE = 500 kcal, then: EEE = TEEE − NEEE = 500 / 45 − ( 2 / 3) ∗ 2.3 = 11.1 − 1.5 = 9.6 kcal / kgFFM For such brief, high-intensity exercise, NEEE (1.5 kcal/kgFFM) is too small to cause an error in judgment about the adequacy of EA. However, if the same TEEE had been expended in 4 hours of gymnastics training, NEEE (9.2 kcal/kgFFM) would be too large to ignore: EEE = TEEE − NEEE = 500 / 45 − 4 ∗ 2.3 = 11.1 − 9.2 = 1.9 kcal / kgFFM If this gymnast were to restrict her dietary intake to EI = 1575 kcal/day, ignoring NEEE would lead to excessive concern about her EA and unwarranted demands for behavior modifications: With NEEE: EA = ( EI − EEE ) / FFM = 1575 / 45 − 1.9 = 33.1 kcal / kgFFM / day (138 kJ / kgFFM / day ) Ignoring NEEE: (EA = EI − TEEE ) / FFM = 1575 / 45 − 9.6 = 25.4 kcal / kgFFM / day (106 kJ / kgFFM / day ) Other sources of error in the calculation of EA derive from errors in the estimation of EI, EEE, and FFM. As pointed out in another review [126], a few simple calculations with realistic values quickly reveal that the greatest efforts should be made to record EI accurately. Consider an athlete with body mass = 60 kg, %Fat = 25%, EEE = 500 kcal/day, and EI = 2100 kcal/day (8.8 MJ/day). Her FFM is (1–0.25) × 60 = 45 kg and her EA is EA = ( EI − EEE ) / FFM = ( 2100 − 500 ) / 45 = 35.6 kcal / kgFFM / day (149 kJ / kgFFM / day ) A 2% error rate in %Fat determinations is not uncommon with body composition analyzers. Subsequently, a 2% overestimate of %Fat (i.e., 27% in the above example) leads to an underestimate of FFM (43.8 kg) and a negligible error in EA: EA = ( 2100 − 500 ) / 43.8 = 36.5 kcal / kgFFM / day (153 kJ / kgFFM / day ) A 10% error in EEE would correspond to a runner erring by half a mile in the length of a 5-mile run. A 10% underestimation of EEE leads to a similarly negligible error in EA: EA = ( 2100 − 450 ) / 45 = 36.7 kcal / kgFFM / day (153 kJ / kgFFM / day ) Underestimations of EI as big as 20% have been suspected by some dietitians. A 20% underestimation of EI would lead to a large error in EA: EA = ( 0.8 × 2100 − 500 ) / 45 = 26.2 kcal / kgFFM / day (110 kJ / kgFFM / day ) Even a 10% underestimation of EI would lead to a substantial error in EA: EA = ( 0.9 × 2100 − 500 ) / 45 = 30.9 kcal / kgFFM / day (129 kJ / kgFFM / day ) 11 Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function 187 A 10% error in EI (210 kcal) is similar to the References energy content of 2–3 slices of bread. If EI is 1. Nattiv A, Loucks AB, Manore MM, Sundgot-Borgen underestimated by 10–20%, then these substanJ, Warren MP. American College of Sports Medicine tial errors in EA will lead to misinterpretations Position Stand. The Female Athlete Triad. Med Sci of experimental data and mismanagement of Sports Exerc. 2007;39(10):1867–82. 2. Otis CL, Drinkwater B, Johnson M, Loucks A, athletes. Therefore, accurate estimations of EA Wilmore J. American College of Sports Medicine depend most importantly on complete dietary position stand. The Female Athlete Triad. Med Sci records. In the Excalibur experiments, the accuSports Exerc. 1997;29(5):i–ix. racy of EI treatments was achieved by adminis3. De Souza MJ, Williams NI, Nattiv A, Joy E, Misra M, Loucks AB, et al. Misunderstanding the Female tering and supervising known meals. Difficult Athlete Triad: refuting the IOC consensus statement as that is for investigators and participants alike, on Relative Energy Deficiency in Sport (RED-S). Br J quantifying EI in observational studies of free-­ Sports Med. 2014;48(20):1461–5. living athletes is even more challenging. 4. Joy E, De Souza MJ, Nattiv A, Misra M, Williams Conclusion: Needed Research 5. More short-term experiments are needed to resolve the ambiguity about whether LH pulsatility depends on energy in general or on specific macronutrients in particular. Clinical trials are needed to verify that women can prevent or reverse functional hypothalamic amenorrhea by dietary reform alone without moderating the exercise regimen and to develop effective interventions that may be sport-specific. In addition, more animal experiments using the new modified activity stress paradigm ([90]) are needed to explore the physiological and neuroendocrine mechanisms of the Female Athlete Triad in more detail. Finally, more experiments like Excalibur III are needed to determine whether other stressors besides exercise have any suppressive effect on LH pulsatility beyond the impact of their energy cost on energy availability. Long-term experiments like Excalibur V are needed to look at EA threshold effects on other aspects of reproductive function besides LH pulsatility, but the expense and controls needed to properly conduct such studies make them challenging to conduct. 6. 7. 8. 9. 10. 11. 12. 13. 14. Conflict of Interest Anne Loucks is a founder and shareholder of AEIOU Scientific, LLC. NI, Mallinson RJ, et al. 2014 Female Athlete Triad Coalition consensus statement on treatment and return to play of the Female Athlete Triad. Curr Sports Med Rep. 2014;13(4):219–32. Mountjoy M, Sundgot-Borgen J, Burke L, Carter S, Constantini N, Lebrun C, et al. The IOC consensus statement: beyond the Female Athlete Triad–Relative Energy Deficiency in Sport (RED-S). Br J Sports Med. 2014;48(7):491–7. Frisch RE, McArthur JW. Menstrual cycles: fatness as determinant of minimum weight for height necessary for their maintenance or onset. Science. 1974;185:949–51. Frisch RE, Revelle R. Height and weight at menarche and a hypothesis of menarche. Arch Dis Child. 1971;46(249):695–701. Schneider JE, Wade GN. Control of fertility by metabolic cues – reply. Am J Physiol Endocrinol Metab. 1997;273(1):E231–E2. Crist DM, Hill JM. Diet and insulin like growth factor I in relation to body composition in women with exercise-induced hypothalamic amenorrhea. J Am Coll Nutr. 1990;9(3):200–4. Bronson FH, Manning JM. The energetic regulation of ovulation: a realistic role for body fat. Biol Reprod. 1991;44(6):945–50. Loucks AB, Horvath SM. Athletic amenorrhea: a review. Med Sci Sports Exerc. 1985;17(1):56–72. Scott EC, Johnston FE. Critical fat, menarche, and the maintenance of menstrual cycles: a critical review. J Adolesc Health Care. 1982;2(4):249–60. Sinning WE, Little KD. Body composition and menstrual function in athletes. Sports Med. 1987;4(1):34–45. Loucks AB, Horvath SM, Freedson PS. Menstrual status and validation of body fat prediction in athletes. Hum Biol. 1984;56(2):383–92. A. B. Loucks 188 15. Bronson FH. Food-restricted, prepubertal, female rats: rapid recovery of luteinizing hormone pulsing with excess food, and full recovery of pubertal development with gonadotropin-releasing hormone. Endocrinology. 1986;118(6):2483–7. 16. Di Carlo C, Palomba S, De Fazio M, Gianturco M, Armallino M, Nappi C. Hypogonadotropic hypogonadotropism in obese women after biliopancreatic diversion. Fertil Steril. 1999;72(5):905–9. 17. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372(6505):425–32. 18. Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, et al. Serum immunoreactive-­ leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334(5):292–5. 19. Tena-Sempere M. Roles of ghrelin and leptin in the control of reproductive function. Neuroendocrinology. 2007;86(3):229–41. 20. Castellano JM, Bentsen AH, Mikkelsen JD, Tena-­ Sempere M. Kisspeptins: bridging energy homeostasis and reproduction. Brain Res. 2010;1364:129–38. 21. Chou SH, Chamberland JP, Liu X, Matarese G, Gao C, Stefanakis R, et al. Leptin is an effective treatment for hypothalamic amenorrhea. Proc Natl Acad Sci U S A. 2011;108(16):6585–90. 22. Ma Z, Gingerich RL, Santiago JV, Klein S, Smith CH, Landt M. Radioimmunoassay of leptin in human plasma. Clin Chem. 1996;42(6 Pt 1):942–6. 23. Laughlin GA, Yen SSC. Hypoleptinemia in women athletes: absence of a diurnal rhythm with amenorrhea. J Clin Endocrinol Metab. 1997;82(1):318–21. 24. Mantzoros C, Flier JS, Lesem MD, Brewerton TD, Jimerson DC. Cerebrospinal fluid leptin in anorexia nervosa: correlation with nutritional status and potential role in resistance to weight gain. J Clin Endocrinol Metab. 1997;82(6):1845–51. 25. Miller KK, Parulekar MS, Schoenfeld E, Anderson E, Hubbard J, Klibanski A, et al. Decreased leptin levels in normal weight women with hypothalamic amenorrhea: the effects of body composition and nutritional intake. J Clin Endocrinol Metab. 1998;83(7):2309–12. 26. Jimerson DC, Mantzoros C, Wolfe BE, Metzger ED. Decreased serum leptin in bulimia nervosa. J Clin Endocrinol Metab. 2000;85(12):4511–4. 27. Hilton LK, Loucks AB. Low energy availability, not exercise stress, suppresses the diurnal rhythm of leptin in healthy young women. Am J Physiol Endocrinol Metab. 2000;278(1):E43–9. 28. Loucks AB, Thuma JR. Luteinizing hormone pulsatility is disrupted at a threshold of energy availability in regularly menstruating women. J Clin Endocrinol Metab. 2003;88(1):297–311. 29. Loucks AB. The response of luteinizing hormone pulsatility to five days of low energy availability disappears by 14 years of gynecological age. J Clin Endocrinol Metab. 2006;91:3158–64. 30. Berga SL, Marcus MD, Loucks TL, Hlastala S, Ringham R, Krohn MA. Recovery of ovarian activity 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. in women with functional hypothalamic amenorrhea who were treated with cognitive behavior therapy. Fertil Steril. 2003;80(4):976–81. Kolaczynski JW, Considine RV, Ohannesian J, Marco C, Opentanova I, Nyce MR, et al. Responses of leptin to short-term fasting and refeeding in humans: a link with ketogenesis but not ketones themselves. Diabetes. 1996;45(11):1511–5. Kolaczynski JW, Ohannesian JP, Considine RV, Marco CC, Caro JF. Response of leptin to short-­ term and prolonged overfeeding in humans. J Clin Endocrinol Metab. 1996;81:4162–5. Weigle DS, Duell PB, Connor WE, Steiner RA, Soules MR, Kuijper JL. Effect of fasting, refeeding, and dietary fat restriction on plasma leptin levels. J Clin Endocrinol Metab. 1997;82:561–5. Jenkins AB, Markovic TP, Fleury A, Campbell LV. Carbohydrate intake and short-term regulation of leptin in humans. Diabetologia. 1997;40(3):348–51. Wang J, Liu R, Hawkins M, Barzilai N, Rossetti L. A nutrient-sensing pathway regulates leptin gene expression in muscle and fat. Nature. 1998;393(6686):684–8. Warren MP. The effects of exercise on pubertal progression and reproductive function in girls. J Clin Endocrinol Metab. 1980;51:1150–7. Winterer J, Cutler GB Jr, Loriaux DL. Caloric balance, brain to body ratio, and the timing of menarche. Med Hypotheses. 1984;15:87–91. Wade GN, Schneider JE. Metabolic fuels and reproduction in female mammals. Neurosci Biobehav Rev. 1992;16(2):235–72. Bronson FH, Heideman PD. Seasonal regulation of reproduction in mammals. In: Knobil E, Neill J, editors. The physiology of reproduction, vol. 2. New York: Raven Press; 1994. p. 541–83. Bronson FH, Manning J. Food consumption, prolonged exercise, and LH secretion in the peripubertal female rat. In: Pirke KM, Wuttle W, Schweiger U, editors. The menstrual cycle and its disorders. Berlin: Springer-Verlag; 1989. p. 42–9. Wade GN, Schneider JE, Li HY. Control of fertility by metabolic cues. Am J Phys. 1996;270(1 Pt 1):E1–19. Furman M, Wade GN. Animal models in the study of nutritional infertility. Curr Opin Endocrinol. 2007;14(6):475–81. Laughlin GA, Yen SSC. Nutritional and endocrine-­ metabolic aberrations in amenorrheic athletes. J Clin Endocrinol Metab. 1996;81(12):4301–9. Christo K, Cord J, Mendes N, Miller KK, Goldstein MA, Klibanski A, et al. Acylated ghrelin and leptin in adolescent athletes with amenorrhea, eumenorrheic athletes and controls: a cross-sectional study. Clin Endocrinol. 2008;69(4):628–33. Scheid JL, Williams NI, West SL, VanHeest SL, De Souza MJ. Elevated PYY is associated with energy deficiency and indices of subclinical disordered eating in exercising women with hypothalamic amenorrhea. Appetite. 2009;52(1):184–92. Myerson M, Gutin B, Warren MP, May MT, Contento I, Lee M, et al. Resting metabolic rate and energy bal- 11 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function ance in amenorrheic and eumenorrheic runners. Med Sci Sports Exerc. 1991;23:15–22. Loucks AB, Laughlin GA, Mortola JF, Girton L, Nelson JC, Yen SSC. Hypothalamic-pituitary-­ thyroidal function in eumenorrheic and amenorrheic athletes. J Clin Endocrinol Metab. 1992;75(2):514–8. Loucks AB, Mortola JF, Girton L, Yen SS. Alterations in the hypothalamic-pituitary-­ ovarian and the hypothalamic-­ pituitary-adrenal axes in athletic women. J Clin Endocrinol Metab. 1989;68(2):402–11. Drinkwater BL, Nilson K, Chesnut CH 3rd, Bremner WJ, Shainholtz S, Southworth MB. Bone mineral content of amenorrheic and eumenorrheic athletes. N Engl J Med. 1984;311(5):277–81. Kaiserauer S, Snyder AC, Sleeper M, Zierath J. Nutritional, physiological, and menstrual status of distance runners. Med Sci Sports Exerc. 1989;21(2):120–5. Marcus R, Cann C, Madvig P, Minkoff J, Goddard M, Bayer M, et al. Menstrual function and bone mass in elite women distance runners. Endocrine and metabolic features. Ann Intern Med. 1985;102(2):158–63. Nelson ME, Fisher EC, Catsos PD, Meredith CN, Turksoy RN, Evans WJ. Diet and bone status in amenorrheic runners. Am J Clin Nutr. 1986;43(6):910–6. Edwards JE, Lindeman AK, Mikesky AE, Stager JM. Energy balance in highly trained female endurance runners. Med Sci Sports Exerc. 1993;25(12):1398-404. Wilmore JH, Wambsgans KC, Brenner M, Broeder CE, Paijmans I, Volpe JA, et al. Is there energy conservation in amenorrheic compared with eumenorrheic distance runners? J Appl Physiol. 1992;72(1):15–22. Mertz W, Tsui JC, Judd JT, Reiser S, Hallfrisch J, Morris ER, et al. What are people really eating? The relation between energy intake derived from estimated diet records and intake determined to maintain body weight. Am J Clin Nutr. 1991;54(2):291–5. Leibel RL, Rosenbaum M, Hirsch J. Changes in energy expenditure resulting from altered body weight. N Engl J Med. 1995;332(10):621–8. Rivier C, Rivest S. Effect of stress on the activity of the hypothalamic-pituitary-gonadal axis: peripheral and central mechanisms. Biol Reprod. 1991;45:523–32. Selye H. The effect of adaptation to various damaging agents on the female sex organs in the rat. Endocrinology. 1939;25:615–24. Asahina K, Kitahara F, Yamanaka M, Akiba T. Influence of excessive exercise on the structure and function of rat organs. Jpn J Physiol. 1959;9:322–6. Axelson JF. Forced swimming alters vaginal estrous cycles, body composition, and steroid levels without disrupting lordosis behavior or fertility in rats. Physiol Behav. 1987;41(5):471–9. Chatterton RT Jr, Hartman AL, Lynn DE, Hickson RC. Exercise-induced ovarian dysfunction in the rat. Proc Soc Exp Biol Med. 1990;193(3):220–4. Manning JM, Bronson FH. Effects of prolonged exercise on puberty and luteinizing hormone secretion in female rats. Am J Phys. 1989;257(6 Pt 2):R1359–64. 189 63. Manning JM, Bronson FH. Suppression of puberty in rats by exercise: effects on hormone levels and reversal with GnRH infusion. Am J Phys. 1991;260(4 Pt 2):R717–23. 64. De Souza MJ, Maguire MS, Maresh CM, Kraemer WJ, Rubin KR, Loucks AB. Adrenal activation and the prolactin response to exercise in eumenorrheic and amenorrheic runners. J Appl Physiol. 1991;70(6):2378–87. 65. De Souza MJ, Luciano AA, Arce JC, Demers LM, Loucks AB. Clinical tests explain blunted cortisol responsiveness but not mild hypercortisolism in amenorrheic runners. J Appl Physiol. 1994;76(3):1302–9. 66. Ding J-H, Scheckter CB, Drinkwater BL, Soules MR, Bremner WJ. High serum cortisol levels in exercise-associated amenorrhea. Ann Intern Med. 1988;108:530–4. 67. Suh BY, Liu JH, Berga SL, Quigley ME, Laughlin GA, Yen SS. Hypercortisolism in patients with functional hypothalamic-amenorrhea. J Clin Endocrinol Metab. 1988;66(4):733–9. 68. Gold PW, Gwirtsman H, Averinos PC, Nieman LK, Gallucci WT, Kaye W, et al. Abnormal hypothalamic-­ pituitary-adrenal function in anorexia nervosa: pathophysiologic mechanisms in underweight and weight-corrected patients. N Engl J Med. 1986;314(21):1335–42. 69. Kuo T, Harris CA, Wang JC. Metabolic functions of glucocorticoid receptor in skeletal muscle. Mol Cell Endocrinol. 2013;380(1–2):79–88. 70. Slentz CA, Davis JM, Settles DL, Pate RR, Settles SJ. Glucose feedings and exercise in rats: glycogen use, hormone responses, and performance. J Appl Physiol. 1990;69:989–94. 71. Tabata I, Ogita F, Miyachi M, Shibayama H. Effect of low blood glucose on plasm CRF, ACTH, and cortisol during prolonged physical exercise. J Appl Physiol. 1991;71:1807–12. 72. Bonen A. Recreational exercise does not impair menstrual cycles: a prospective study. Int J Sports Med. 1992;13(2):110–20. 73. Boyden TW, Pamenter RW, Stanforth P, Rotkis T, Wilmore JH. Sex steroids and endurance running in women. Fertil Steril. 1983;39(5):629–32. 74. Bullen BA, Skrinar GS, Beitins IZ, Carr DB, Reppert SM, Dotson CO, et al. Endurance training effects on plasma hormonal responsiveness and sex hormone excretion. J Appl Physiol. 1984;56(6):1453–63. 75. Rogol AD, Weltman JY, Evans WS, Veldhuis JD, Weltman AL. Long-term endurance training alters the hypothalamic-pituitary axes for gonadotropins and growth hormone. Endocrinol Metab Clin North Am. 1992;21(4):817–32. 76. Loucks AB, Cameron JL, De Souza MJ. Subject assignment may have biased exercise results [letter; comment]. J Appl Physiol. 1993;74(4):2045–7. 77. Bullen BA, Skrinar GS, Beitins IZ, von Mering G, Turnbull BA, McArthur JW. Induction of menstrual disorders by strenuous exercise in untrained women. N Engl J Med. 1985;312(21):1349–53. 190 78. Williams NI, Young JC, McArthur JW, Bullen B, Skrinar GS, Turnbull B. Strenuous exercise with caloric restriction: effect on luteinizing hormone secretion. Med Sci Sports Exerc. 1995;27(10):1390–8. 79. Loucks AB, Callister R. Induction and prevention of low-T3 syndrome in exercising women. Am J Phys. 1993;264(5 Pt 2):R924–30. 80. Loucks AB, Heath EM. Induction of low-T3 syndrome in exercising women occurs at a threshold of energy availability. Am J Phys. 1994;266(3 Pt 2): R817–23. 81. Loucks AB, Heath EM. Dietary restriction reduces luteinizing hormone (LH) pulse frequency during waking hours and increases LH pulse amplitude during sleep in young menstruating women. J Clin Endocrinol Metab. 1994;78(4):910–5. 82. Loucks AB, Verdun M, Heath EM. Low energy availability, not stress of exercise, alters LH pulsatility in exercising women. J Appl Physiol. 1998;84(1): 37–46. 83. Loucks AB, Verdun M. Slow restoration of LH pulsatility by refeeding in energetically disrupted women. Am J Phys. 1998;275(4 Pt 2):R1218–26. 84. Loucks A. Is stress measured in joules? Mil Psychol. 2009;21(S1):S101–S7. 85. Treloar AE, Boynton RE, Behn BG, Brown BW. Variation of the human menstrual cycle through reproductive life. Int J Fertil. 1967;12(1 Pt 2):77–126. 86. Olson BR, Cartledge T, Sebring N, Defensor R, Nieman L. Short-term fasting affects luteinizing hormone secretory dynamics but not reproductive function in normal-weight sedentary women. J Clin Endocrinol Metab. 1995;80(4):1187–93. 87. Alvero R, Kimzey L, Sebring N, Reynolds J, Loughran M, Nieman L, et al. Effects of fasting on neuroendocrine function and follicle development in lean women. J Clin Endocrinol Metab. 1998;83(1): 76–80. 88. Williams NI, Caston-Balderrama AL, Helmreich DL, Parfitt DB, Nosbisch C, Cameron JL. Longitudinal changes in reproductive hormones and menstrual cyclicity in cynomolgus monkeys during strenuous exercise training: abrupt transition to exercise-induced amenorrhea. Endocrinology. 2001;142(6):2381–9. 89. Williams NI, Helmreich DL, Parfitt DB, Caston-­ Balderrama AL, Cameron JL. Evidence for a causal role of low energy availability in the induction of menstrual cycle disturbances during strenuous exercise training. J Clin Endocrinol Metab. 2001;86(11):5184–93. 90. DiMarco NM, Dart L, Sanborn C. Modified activity-­ stress paradigm in an animal model of the Female Athlete Triad. J Appl Physiol. 2007;103:1469–78. 91. Bursztein S, Elwyn DH, Askanazi J, Kinney JM. Fuel utilization in normal, starving, and pathological states. Energy metabolism, indirect calorimetry, and nutrition. Baltimore: Williams & Wilkins; 1989. p. 146. 92. Bronson FH, Heideman PD. Short-term hormonal responses to food intake in peripubertal female rats. Am J Phys. 1990;259(1 Pt 2):R25–31. A. B. Loucks 93. Foster DL, Ebling FJ, Micka AF, Vannerson LA, Bucholtz DC, Wood RI, et al. Metabolic interfaces between growth and reproduction. I. Nutritional modulation of gonadotropin, prolactin, and growth hormone secretion in the growth-limited female lamb. Endocrinology. 1989;125(1):342–50. 94. McCann JP, Hansel W. Relationships between insulin and glucose metabolism and pituitary-­ ovarian functions in fasted heifers. Biol Reprod. 1986;34(4):630–41. 95. Parfitt DB, Church KR, Cameron JL. Restoration of pulsatile luteinizing hormone secretion after fasting in rhesus monkeys (Macaca mulatta): dependence on size of the refeed meal. Endocrinology. 1991;129(2):749–56. 96. Schreihofer DA, Renda F, Cameron JL. Feeding-­ induced stimulation of luteinizing hormone secretion in male rhesus monkeys is not dependent on a rise in blood glucose concentration. Endocrinology. 1996;137(9):3770–6. 97. Flatt JP. Energetics of intermediary metabolism. In: Kinney JM, editor. Assessment of energy metabolism in health and disease. Columbus: Ross Laboratories; 1980. p. 77–87. 98. Loucks AB. Low energy availability in the marathon and other endurance sports. Sports Med. 2007;37(4–5):348–52. 99. Kopp-Woodroffe SA, Manore MM, Dueck CA, Skinner JS, Matt KS. Energy and nutrient status of amenorrheic athletes participating in a diet and exercise training intervention program. Int J Sport Nutr. 1999;9(1):70–88. 100. De Souza MJ, Miller BE, Loucks AB, Luciano AA, Pescatello LS, Campbell CG, et al. High frequency of luteal phase deficiency and anovulation in recreational women runners: blunted elevation in follicle-stimulating hormone observed during luteal-follicular transition. J Clin Endocrinol Metab. 1998;83(12):4220–32. 101. Ihle R, Loucks AB. Dose-response relationships between energy availability and bone turnover in young exercising women. J Bone Mineral Res. 2004;19(8):1231–40. 102. Compston JE. Sex steroids and bone. Physiol Rev. 2001;81(1):419–47. 103. Baker ER, Mathur RS, Kirk RF, Williamson HO. Female runners and secondary amenorrhea: correlation with age, parity, mileage, and plasma hormonal and sex-hormone-binding globulin concentrations. Fertil Steril. 1981;36(2):183–7. 104. Ellison PT. Advances in human reproductive ecology. Annu Rev Anthropol. 1994;23:255–75. 105. Weaver CM, Martin BR, Plawecki KL, Peacock M, Wood OB, Smith DL, et al. Differences in calcium metabolism between adolescent and adult females. Am J Clin Nutr. 1995;61(3):577–81. 106. Williams NI, Reed JL, Leidy HJ, Legro RS, De Souza MJ. Estrogen and progesterone exposure is reduced in response to energy deficiency in women aged 25–40 years. Hum Reprod. 2010;25(9):2328–39. 11 Exercise Training in the Normal Female: Effects of Low Energy Availability on Reproductive Function 107. Lieberman JL, DS MJ, Wagstaff DA, Williams NI. Menstrual disruption with exercise is not linked to an energy availability threshold. Med Sci Sports Exerc. 2018;50(3):551–61. 108. Williams NI, Leidy HJ, Hill BR, Lieberman JL, Legro RS, De Souza MJ. Magnitude of daily energy deficit predicts frequency but not severity of menstrual disturbances associated with exercise and caloric restriction. Am J Physiol Endocrinol Metab. 2015;308(1):E29–39. 109. Redman LM, Loucks AB. Menstrual disorders in athletes. Sports Med. 2005;35(9):747–55. 110. Vollman RF. The menstrual cycle. Major Probl Obstet Gynecol. 1977;7:1–193. 111. Cialdella-Kam L, Guebels CP, Maddalozzo GF, Manore MM. Dietary intervention restored menses in female athletes with exercise-associated menstrual dysfunction with limited impact on bone and muscle health. Nutrients. 2014;6(8):3018–39. 112. Guebels CP, Kam LC, Maddalozzo GF, Manore MM. Active women before/after an intervention designed to restore menstrual function: resting metabolic rate and comparison of four methods to quantify energy expenditure and energy availability. Int J Sport Nutr Exerc Metab. 2014;24(1): 37–46. 113. Dueck CA, Matt KS, Manore MM, Skinner JS. Treatment of athletic amenorrhea with a diet and training intervention program. Int J Sport Nutr. 1996;6(1):24–40. 114. Loucks AB, Kiens B, Wright HH. Energy availability in athletes. J Sport Sci. 2011;S1:S7–S15. 115. Braun DL, Sunday SR, Halmi KA. Psychiatric comorbidity in patients with eating disorders. Psychol Med. 1994;24(4):859–67. 116. Kaye WH, Bulik CM, Thornton L, Barbarich N, Masters K. Comorbidity of anxiety disorders with anorexia and bulimia nervosa. Am J Psychiat. 2004;161(12):2215–21. 191 117. Loucks AB. Energy balance and body composition in sports and exercise. J Sport Sci. 2004;22:1–14. 118. Thomas DT, Erdman KA, Burke LM. American College of Sports Medicine joint position statement. Nutrition and athletic performance. Med Sci Sports Exerc. 2016;48(3):543–68. 119. Hubert P, King NA, Blundell JE. Uncoupling the effects of energy expenditure and energy intake: appetite response to short-term energy deficit induced by meal omission and physical activity. Appetite. 1998;31(1):9–19. 120. Blundell JE, King NA. Physical activity and regulation of food intake: current evidence. Med Sci Sports Exerc. 1999;31(11 Suppl):S573–83. 121. Stubbs RJ, Hughes DA, Johnstone AM, Whybrow S, Horgan GW, King N, et al. Rate and extent of compensatory changes in energy intake and expenditure in response to altered exercise and diet composition in humans. Am J Phys. 2004;286(2):R350–8. 122. Horvath PJ, Eagen CK, Ryer-Calvin SD, Pendergast DR. The effects of varying dietary fat on the nutrient intake in male and female runners. J Am Coll Nutr. 2000;19(1):42–51. 123. Horvath PJ, Eagen CK, Fisher NM, Leddy JJ, Pendergast DR. The effects of varying dietary fat on performance and metabolism in trained male and female runners. J Am Coll Nutr. 2000;19(1):52–60. 124. Wardle J, Haase AM, Steptoe A. Body image and weight control in young adults: international comparisons in university students from 22 countries. Int J Obesity. 2006;30(4):644–51. 125. Martinsen M, Bratland-Sanda S, Eriksson AK, Sundgot-Borgen J. Dieting to win or to be thin? A study of dieting and disordered eating among adolescent elite athletes and non-athlete controls. Br J Sport Med. 2010;44(1):70–6. 126. Loucks AB. The Female Athlete Triad: a metabolic phenomenon. Pensar En Movimiento. 2014;12(1): 1–23. Ghrelin Responses to Acute Exercise and Training 12 Jaak Jürimäe Introduction The importance of physical exercise to influence energy balance and body mass is widely recognized [1]. A complex neuroendocrine system is involved in the regulation of energy homeostasis including central and peripheral tissues [2, 3]. Important to this regulatory system is the existence of several appetite hormones, including adipose and gut tissue hormones that communicate the status of body energy stores to the hypothalamus [2]. Energy intake is an integral to energy balance and is regulated via neuronal circuits interacting with gut hormones, key among these being ghrelin and peptide YY [4, 5]. It appears that peptide YY functions as a negative feedback signal and is responsible for inducing satiety and cessation of eating after food intake [5]. In contrast, ghrelin is a hormone well known for its acute orexigenic properties stimulating food consumption [6, 7]. Changes in these circulating appetite hormones influence the physiological drive to eat, weight gain and also reproductive function [4]. Furthermore, ghrelin may also be involved in pubertal development, where rapid growth and development need careful coordination of energy balance and appetite regulatory signals [4]. Finally, circulating ghrelin concentraJ. Jürimäe (*) Institute of Sport Sciences and Physiotherapy, University of Tartu, Tartu, Estonia e-mail: jaak.jurimae@ut.ee tions may vary dramatically depending on specific body composition, physical activity and physical fitness parameters [2]. This chapter focuses on the available information about the effects of acute exercise and chronic exercise training on the secretion of ghrelin. Ghrelin, a peptide secreted by distinct endocrine cells of the stomach, was first described as an endogenous ligand for the growth hormone secretagogue receptor [8]. However, ghrelin role in body mass regulation is more prominent than its role in growth hormone secretion [9]. Ghrelin promotes positive energy balance by increasing appetite and food intake [10, 11]. Specifically, the rise in circulating ghrelin concentration before a meal is a physiological signal for hunger and the body’s cue for meal initiation [12]. Therefore, the rise in ghrelin levels and hunger occurs independent of food and time of day cues [12]. Meal responses of ghrelin are related to acute caloric intake over a typical day of eating in normal-weight subjects [13]. Furthermore, ghrelin levels have been demonstrated to be negatively correlated with 24-h caloric intake [14], and ghrelin concentrations decrease after caloric intake and increase while fasting [2]. The decrease in ghrelin release is related to the specific amount of calories ingested [15]. Accordingly, ghrelin is responsive to diet- and exercise-induced changes in body mass [16]. In addition to total ghrelin, acylated and des-­ acylated forms of ghrelin have been described © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_12 193 194 [17]. The acylated form of ghrelin is thought to be essential for ghrelin biological activity [18], whereas unacylated ghrelin has been suggested to be biologically inactive [19]. Specifically, acylated ghrelin has been reported to be associated with the regulation of growth hormone secretion, cardiac performance, cell proliferation and adipogenesis and affects appetite, food intake and energy balance [8, 20, 21]. There are also some studies suggesting that unacylated ghrelin is related to insulin resistance [22–24]. It has also been demonstrated that total ghrelin and acylated ghrelin are positively correlated [25–27] and both forms of the ghrelin potentially play a role in energy balance [28]. Based on these results, it could be suggested that acylated and desacylated forms of ghrelin change similar to changes in energy balance, and total ghrelin concentration can be used as a biomarker in energy balance studies [4, 29, 30]. Future studies, nonetheless, are needed to better clarify the responses of total ghrelin and its specific forms in various conditions of energy balance. hrelin During Growth G and Maturation in Children Ghrelin is a hormone that could influence somatic growth [4] and sexual maturation [31]. Specifically, a negative association of circulating ghrelin level with age [4, 31] and pubertal development [32] has been found. It has been hypothesized that ghrelin provides a link between energy homeostasis, body composition and pubertal development through actions on the hypothalamus [33], where ghrelin stimulates the secretion of gonadotropin-releasing hormone, which in turn stimulates the secretion of the gonadotropins required for pubertal onset [16]. It has been found that the initiation of puberty substantially decreases ghrelin concentrations in both sexes [4, 31]. A negative correlation between ghrelin and testosterone has been found in boys entering puberty [32]. In contrast, a recent study demonstrated no effect of testosterone and estradiol on ghrelin decrease during pubertal growth in boys and girls, respectively J. Jürimäe [4]. It was found that a drop in circulating total ghrelin to its lowest levels occurred during peak pubertal growth [4]. Furthermore, Cheng et al. [4] suggested that adolescent ghrelin concentrations may be more strongly associated with markers of somatic growth than sexual maturation. Specifically, circulating ghrelin levels were inversely correlated with insulin-like growth factor-1 concentrations and with annual height and weight velocity in both sexes [4]. Accordingly, the decrease in circulating ghrelin levels at the onset of puberty is apparent [4, 34, 35], despite the fact that puberty is characterized by increased appetite and food intake [31] and ghrelin is known to stimulate appetite [28, 36]. Research suggests that there could be an increased sensitivity for appetite stimulation by ghrelin over puberty [31] and/or low ghrelin concentrations signal adequate nutritional status to support rapid somatic growth and development of reproductive capacity [4] to sustain growth in this period. In addition, elevated energy expenditure and, therefore, also an increased energy intake in physically active children during pubertal maturation are linked to higher circulating ghrelin levels in these children compared with physically inactive children [36]. Accordingly, it could be argued that regular physical activity still causes higher ghrelin levels during puberty to stimulate appetite and food intake to cover higher energy homeostasis [36]. This is supported by the finding that there is a negative correlation of cardiorespiratory fitness as measured by peak oxygen consumption with total ghrelin [37] and acylated and desacylated forms of ghrelin [38] in boys during puberty. However, different forms of ghrelin were not associated with directly measured physical activity intensities in pubertal boys with differing body composition [38]. Collectively, these results demonstrate that somatic growth and maturation are associated with ghrelin, which concentrations decrease with advancing age and puberty. However, further longitudinal studies throughout puberty in children with various physical activity and body composition levels are needed to better understand how physical fitness and activity may influence circulating ghrelin concentrations dur- 12 Ghrelin Responses to Acute Exercise and Training ing puberty in children with different body ­composition values before any definitive conclusions can be drawn. In longitudinal investigations with growing and maturing athletes, total ghrelin levels have been studied in female gymnasts [35, 39] and male and female swimmers [40, 41]. It could be argued that regular sport training increases ghrelin levels to stimulate appetite and food intake to cover higher energy homeostasis in these young athletes [2, 42]. Ghrelin may act as a hormone signalling a need for energy conservation, and ghrelin secretion is triggered to counter a further deficit in energy storage to help to maintain body mass [2, 43]. Accordingly, higher basal ghrelin concentrations have been found in prepubertal and adolescent athletes when compared with untrained controls [34, 35, 44]. However, basal ghrelin levels decreased in both prepubertal rhythmic gymnasts and age-matched lean untrained controls over a 12-month study period [39], showing that an increasing age decreases ghrelin concentrations similarly in both groups despite large differences in daily energy expenditure [2, 36]. Therefore, ghrelin concentrations were still significantly higher in the rhythmic gymnasts when compared with untrained controls at both measurement times during prepuberty [39]. However, when rhythmic gymnasts and untrained controls reached puberty, ghrelin levels were decreased in both groups and were not different between groups with different energy expenditure levels [35]. Similarly, a significant decrease in basal ghrelin levels was observed in male swimmers after the evolution of puberty [41], while basal ghrelin levels were not changed in pubertal female swimmers with advancing pubertal maturation over a 2-year study period [40]. It can be suggested that basal ghrelin levels are higher in prepubertal children who participate in sport training in comparison with age-matched untrained controls, while basal ghrelin levels decrease when young athletes reach puberty even in the presence of chronically elevated energy expenditure [2, 36]. Furthermore, pubertal maturation appears to reduce circulating ghrelin concentrations in growing athletes of both sexes, despite heavy athletic activity [2, 36]. 195 hrelin Relationships with G Adiposity and Energy Availability Ghrelin levels are significantly lower in obese individuals [45–47] and substantially elevated in patients with anorexia nervosa [12, 48, 49], proposed as a likely adaptive mechanism response [12, 50]. Accordingly with these patterns, there is a negative association of ghrelin concentration with body mass [32, 51], body mass index [32, 52], total body fat mass [34, 51], visceral fat mass [53, 54] and total body lean mass [32, 55]. It has also been suggested that circulating ghrelin level could be regarded as a signal of decreased total body lean mass in healthy elderly females [56]. In addition, there are also studies to show an inverse correlation between ghrelin concentration and body height [32, 57, 58] and body height velocity [4] during growth in children. Diet-induced weight loss in obese individuals has been accompanied by increases in circulating total ghrelin concentrations [59]. For example, plasma ghrelin levels increased by 17% in overweight women who reduced their body mass by 4.5% after 10-week body weight loss intervention programme [60], while a 6-month supervised weight loss programme that caused 17.4% body weight loss induced 24% increase in ghrelin levels [61]. In addition, short-term diet-­ induced body weight loss in obese subjects resulted in higher total ghrelin concentrations, which remained elevated also over weight maintenance periods of 6 and 12 months [59]. Similarly, long-term exercise intervention together with diet-control investigations has demonstrated that total ghrelin levels increase in response to exercise-induced body weight loss in obese subjects and not because of food restriction per se, acting via a negative feedback loop that regulates body mass [7, 62]. It has been suggested that changes in total ghrelin concentrations appear to be most sensitive to changes in body mass resulting from overall energy deficit, independent of specific effects of nutritional intake and/or physical exercise [7, 62]. There are studies to demonstrate that manipulations in food intake and exercise energy expenditure show a close relationship between circulating ghrelin 196 and energy availability [63, 64]. For example, Scheid et al. [64] measured total ghrelin, energy balance and body composition parameters before and after 3-month intervention period in exercising women and found that circulating ghrelin does not play a role in the adaptive changes associated with exercise training when exercise occurs in the absence of body weight loss. However, fasting ghrelin level increased when body mass is lost and may respond to even smaller changes in energy availability [64]. In addition, the change in total ghrelin level was inversely correlated with the change in body mass, body mass index, lean body mass and energy availability after diet- and exercise-­ associated weight loss [64]. In contrast, no impact of aerobic training on acylated ghrelin levels was observed in overweight and obese men [65]. It has been suggested that differences in body fat mass loss, exercise volume and duration, and gender may influence possible differences in ghrelin responses to weight reduction [59]. In addition, King et al. [66] showed that equivalent energy deficits induced by food restriction or physical exercise have markedly different effects on appetite, energy intake and acylated ghrelin concentrations. While food restriction elicited a rapid increase in appetite and energy intake and these responses appear to be related to postprandial suppression of acylated ghrelin, acute energy deficits induced by vigorous intensity exercise session did not alter appetite or energy intake and may be related to the failure of acute exercise to induce compensatory acylated ghrelin responses [66]. These results together suggest that changes in body mass are needed before any changes in circulating ghrelin levels could be seen in untrained individuals. Ghrelin Responses to Acute Exercise There are a number of studies including athletes that have investigated the influence of acute bout of exercise on total ghrelin [37, 67–82] and on acylated ghrelin [66, 83–98] concentrations. Different investigations with healthy untrained individuals [37, 67, 68, 78] and also well-trained J. Jürimäe endurance athletes [71, 74, 82] would suggest that exercise-induced acute negative energy balance may not be sufficient to alter total ghrelin response. Conversely, however, there are studies demonstrating that total ghrelin level increased [69, 70, 72, 73] or decreased [75–77, 80, 81] as a result of short-term exercise session. In addition, studies with acylated ghrelin have mostly reported significant suppression [83, 84, 86, 89, 90, 94–98] or no change [66, 85, 92] in measured acylated ghrelin concentration after acute exercise. However, there are also studies that have observed significant postexercise increase in acylated ghrelin concentration [91, 93]. Accordingly, acute exercise studies have demonstrated different responses of different ghrelin forms to the acute exercise in subjects with different body composition and physical activity levels. A study by Dall et al. [68] reported no change in total ghrelin concentration after acute cycling exercise for 45 min at the intensity of anaerobic threshold in healthy middle-aged men. Similarly, total ghrelin levels remained unchanged after acute submaximal running workloads (50%, 70% and 90% of maximal oxygen consumption [VO2MAX]) [78] and also after a single bout of treadmill running for 60 min [67] in healthy physically fit male individuals. In well-trained endurance athletes, a progressively intense intermittent exercise trial on treadmill at different exercise intensities (10 min at 60%, 10 min at 75%, 5 min at 90% and 2 min at 100% of VO2MAX [74] and 30 min on-water sculling exercise performed either below or above the intensity of individual anaerobic threshold [71] did not change total ghrelin concentration. It could be argued that acute exercise energy expenditure was not sufficient to alter total ghrelin response in these studies [1]. Accordingly, significant postexercise increases in total ghrelin concentration after prolonged 2-h endurance rowing at the intensity of 80% of individual anaerobic threshold [34] and after 3-h endurance cycling at the intensity of 50% of maximal aerobic power [69] have been observed in endurance-trained athletes. Assuming that the energy balance drives the ghrelin response to prolonged rowing 12 Ghrelin Responses to Acute Exercise and Training e­ xercise with the estimated energy expenditure of 1200–1500 kcal, it was conceivable to see that the increased postexercise total ghrelin concentration was associated with the amount of work performed (r = 0.75; p < 0.05) in rowers [34]. Furthermore, it was argued that the reduced resting levels of total ghrelin may have influenced the significant exercise-induced increase in ghrelin concentration in rowers [34]. The results of these studies [34, 69] would suggest that a certain threshold reduction in energy availability should be reached before any significant postexercise increases in total ghrelin concentration occur and that the amplitude of the total ghrelin increase could be linked to the energetic status induced by acute exercise stress and the resting levels of ghrelin in athletes [1]. However, to what extent exercise intensity may influence total ghrelin response to acute exercise has not yet been determined, although it has been suggested that low- rather than high-intensity exercise with longer duration stimulates total ghrelin levels [70]. Specifically, Erdmann et al. [70] investigated the effect of exercise intensity and duration on total ghrelin release, hunger and food intake in normal-­weight untrained healthy individuals. Total ghrelin concentrations were increased by 50–70 pg/ml as a result of prolonged low-intensity bicycling exercise with a duration of up to 2 h, while no changes in total ghrelin were observed during higher intensity exercise [70]. In addition, only 2-h prolonged aerobic exercise at the intensity of 50 W with an exercise energy expenditure of 340 kcal lead to an increase in food intake without having an effect on hunger sensations [70]. An increase in plasma ghrelin concentration during exercise without alterations of hunger sensations under similar conditions of low-intensity exercise and energy expenditure was also found in another study [79]. Nonetheless, the stimulation of food intake during prolonged exercise was most likely not due to changes in circulating total ghrelin levels [70]. These results together demonstrate that total ghrelin concentrations can be increased as a result of a low-intensity prolonged exercise session when the exercise energy expenditure is high enough also in untrained subjects. 197 There are studies to suggest that acute exercise stress could also result in a decrease of total ghrelin concentration [75, 77, 80, 81]. These studies have used more intensive exercise bouts including resistance exercise protocols [75, 77, 80, 81], and it has been suggested that glucoregulatory stress from the acute intense exercise could result in a suppression of circulating ghrelin during the recovery period from the exercise [74, 75]. Indeed, studies that have utilized more intensive exercise bouts have demonstrated that maximal exercise-induced large increases in insulin [74, 75] and growth hormone [75, 81] levels may suppress total ghrelin concentration during the recovery period. However, there are also investigations that contradict the results of these studies as exercise-induced increases in both total ghrelin and growth hormone values have been observed after prolonged low-intensity exercise in endurance-trained males [69] and also in overweight postmenopausal women [79]. Others have argued that postexercise ghrelin responses may be independent of changes in energy balance [6] and that acute exercise stress increases energy intake only some time postexercise [6, 83]. To this end, Broom et al. [83] investigated the effects of 1 h running at 72% of VO2MAX on total and acylated ghrelin concentrations. They found that total ghrelin was not changed, while acylated ghrelin was decreased as a result of exercise [83]. Accordingly, it has been argued that although there is a close relationship between total and acylated ghrelin concentrations [25–27], it cannot be excluded that after acute exercise this relationship may be somewhat different [42, 70, 83]. Different studies have demonstrated that relatively high-intensity exercise sessions (≥70% VO2MAX) may suppress acylated ghrelin concentrations [99, 100]. Typically, this hormonal decrease coincides with a transient reduction in appetite during and immediately after the exercise [87, 88], while there are also studies that have found no changes in appetite as a result of acute exercise [89, 92, 101]. It is possible that the lack of commonly observed appetite suppression may be due to a difference in training status or fitness of studied subjects [89]. In accordance, there is an evidence to suggest that highly trained J. Jürimäe 198 individuals are more accustomed to exercise stress and therefore do not have as great hormonal, including acylated ghrelin, response to acute exercise as in untrained individuals [88, 99]. For example, Broom et al. [84] found that plasma acylated ghrelin and hunger ratings fell and remained suppressed for 1.5 h after 90 min running at the intensity of 70% of VO2MAX (≈70% decrease in acylated ghrelin) in healthy men. In other studies with endurance-trained men, circulating acylated ghrelin concentrations were decreased after 45 min of cycling at the intensity of ≈76% of VO2MAX (≈23% decrease in acylated ghrelin) [89] and after 20 km run (≈14% decrease in acylated ghrelin) [90]. Therefore, the suppression of acylated ghrelin in endurance-trained athletes was transient, with concentrations not different from baseline already after 30 [90] and 40 [89] min postexercise. A recent study by Mattin et al. [92] observed no significant changes in acylated ghrelin and appetite scores as a result of 60 min cycling at the intensities of 40% and 70% of VO2MAX in healthy men. Therefore, although not statistically significant, acylated ghrelin responded differently to exercise intensity, as serum levels decreased by ≈27% at the intensity of 70% of VO2MAX and increased by ≈12% at the intensity of 40% of VO2MAX [92]. Larson-Meyer et al. [91] also found a significant increase in acylated ghrelin immediately after 60 min running at the intensity of 70% of VO2MAX in female runners. Therefore, appetite was not affected by running exercise, and postexercise acylated ghrelin was not associated with appetite scores [91]. It was argued that the energy cost of the running exercise may promote increased acylated ghrelin secretion after exercise in these athletes [91]. The results also suggested that acylated ghrelin is not a major contributor to postexercise food intake, perhaps because the signal is dampened by increases in different anorexigenic peptides at the same time [91, 102]. In accordance, other studies have also argued that it is possible that the transient suppression of circulating acylated ghrelin that can be observed during acute exercise may be entirely unrelated to appetite regulation [50, 85]. These results together suggest that acylated ghrelin is responsive to dif- ferent conditions and modes of endurance exercise, duration and intensity, but the direction of the hormone response can be varied [95]. The differences in acylated ghrelin responses to acute exercise can also be attributed to subject physical fitness, pre-exercise meal consumption and timing as well as the timing of the hormone measurements and possible environmental factors such as temperature and altitude [103]. There is a need for further investigations to elucidate the exact mechanisms regulating ghrelin synthesis and clearance during and after acute exercise. Chronic Exercise Training and Ghrelin Responses Chronic exercise training perturbs energy balance and can potentially alter body mass and composition. There are a number of studies that have reported an increase in circulating ghrelin concentrations after long-term exercise interventions in previously untrained individuals [13, 43, 62, 104–108], while other studies have not found any changes in ghrelin concentrations as a result of prolonged exercise training [51, 109–111]. It appears that circulating ghrelin levels increase with body weight loss [62, 105, 107, 108] and decrease with body weight gain [12, 112]. Accordingly, data on ghrelin responses to prolonged exercise training are mainly available from obese individuals (i.e. individuals involved in weight loss programme) [62, 106, 107, 109, 113], whereas only limited data are provided for athletes [29, 30, 114–116]. Most of the previous investigations have studied total ghrelin response to prolonged exercise training [13, 29, 30, 43, 62, 108, 109, 114, 115], while relatively few intervention studies have measured acylated [111, 113, 116] or unacylated [23, 24] ghrelin concentrations separately. Currently, there appears to be only one published study that has investigated the response of acylated ghrelin to prolonged training period in athletes [116]. Previous investigations have mostly found that total ghrelin concentrations increase during situations of body weight loss and suggest that weight loss is the most potential factor influencing ghre- 12 Ghrelin Responses to Acute Exercise and Training lin response to exercise training [13, 43, 62, 107, 108, 117]. In an earlier study, Leidy et al. [108] found that fasting ghrelin concentration was increased twofold in a group of normal-weight women who experienced weight loss (>1.5 kg) as a result of a 3-month energy deficit-imposing diet and 5-days-a-week exercise training intervention programme [108]. Therefore, body mass, body fat mass and resting metabolic rate significantly decreased before the increase in fasting ghrelin occurred [108]. It was suggested that circulating total ghrelin responds in a compensatory manner to changes in energy homeostasis in healthy young women and that ghrelin exhibits particular sensitivity to changes in body mass [108]. In another study, Foster-Schubert et al. [62] reported that total ghrelin levels increased by 18% in sedentary overweight postmenopausal women who lost more than 3 kg body mass after 1-year aerobic exercise training programme. Another 1-year moderate-to-vigorous intensity aerobic exercise for 45 min 5 days a week demonstrated that greater weight loss was associated with larger increases in total ghrelin concentrations in overweight and obese postmenopausal women [107]. Similarly, moderate-intensity aerobic exercise training 5 days a week for 12 weeks increased circulating acylated ghrelin concentrations in overweight and obese men and women [113]. In contrast to these findings, fasting acylated ghrelin concentrations decreased after a moderate dose (14 kcal/kg body mass weekly) but did not change after a low-dose (8 kcal/kg body mass weekly) moderate-intensity aerobic exercise training lasting 4 months in healthy nonobese older women [111]. It was argued that exercise training dose can have specific effects on acylated ghrelin that are not dependent on body weight or body fat mass loss [111]. However, there was a lack of acylated ghrelin level change in those participants who lost body weight or body fat mass as a result of 4-month training period [111]. In another study, Ravussin et al. [51] observed that neither positive energy balance caused by overfeeding nor negative energy balance induced by exercise training had a significant effect on total ghrelin concentration over a 100-day study period. The impact of negative energy balance on total ghrelin 199 levels at the end of the investigation was smaller, due to the possible effect of accustomization [51]. Another study with a group of morbidly obese men and women demonstrated that fasting circulating total ghrelin levels remained unchanged despite 5% body weight loss induced by a 3-week integrated body weight reduction programme with exercise training [109]. The amplitude of ghrelin response to negative energy balance in these studies could be linked to the energetic status of studied individuals, which is attributable to specific body fat mass and exercise training characteristics. Accordingly, data regarding the influence of exercise training programme on circulating ghrelin in previously untrained individuals suggests that exercise training per se has no impact on circulating ghrelin levels and changes in ghrelin concentrations that are seen as a result of exercise training intervention take place as secondary changes to body weight loss [117]. Evidence suggests that the degree of negative energy balance and/or body weight loss threshold to increase circulating ghrelin concentrations has not yet been determined [1, 17]. In heavily exercising females, menstrual disturbances have been linked to an energy deficiency, where caloric intake is inadequate for exercise energy expenditure [12, 118]. These menstrual disturbances, together with an energy deficiency, are largely attributable to athletic events, where the emphasis is on the achievement of thin and lean physiques, which may require low body mass and body fat percent such as in gymnastics, figure skating and long-distance running [12]. Accordingly, higher ghrelin levels have been observed in amenorrheic athletes than in normally ovulating women who train [17, 119]. In fact, there are data to suggest that young female athletes with varying severities of menstrual disturbances can be distinguished from each other based on their circulating ghrelin levels [12, 48, 120, 121]. To this end, as energy deficiency increases in severity across the continuum of menstrual cycle disturbances, physically active women with amenorrhea have the lowest resting energy expenditure relative to lean body mass, together with the increased ghrelin levels [48, 121]. In contrast, physically active women with 200 subtle menstrual disturbances and nonathletic controls present higher resting energy expenditure relative to lean body mass and lower ghrelin concentrations [48, 121]. Increased ghrelin levels in young female athletes with amenorrhea may have a role in reproductive system [12, 119, 120]. An inverse relationship between acylated ghrelin concentration and gonadal steroids was observed in athletes [48], and acylated ghrelin levels may differentiate between athletes who will or will not develop functional hypothalamic amenorrhea during heavy training [48, 119]. Accordingly, it is likely that high circulating ghrelin concentrations contribute to functional hypothalamic amenorrhea by altering gonadotropin-releasing hormone and luteinizing hormone pulsatility [119, 120]. Therefore, body fat mass has an important negative influence on basal ghrelin levels in amenorrheic athletes [48, 120]. An increase in energy intake in amenorrheic athletes induces a decrease in basal ghrelin concentrations, which is paralleled by increases in body mass and resumption of menses [119]. Accordingly, it appears that circulating ghrelin is a biomarker of energy imbalance across the menstrual cycle in female athletes [36, 122]. Since ghrelin levels are consistently elevated in energy deficiency such as functional hypothalamic amenorrhea, ghrelin could be an important marker of energy deficiency and chronic undernutrition [12] and should be measured to monitor the health of female athletes. The mechanisms by which changes in energy balance and/or body mass impact on circulating ghrelin levels are not fully understood [2, 36, 117]. It has been proposed that leptin, which levels directly correlate with body fat mass, may have an influence on circulating ghrelin concentrations [117]. Specifically, a negative association between circulating leptin and ghrelin concentrations has been reported [34], and an increase in circulating ghrelin levels in response to body weight loss may therefore occur as a result of a decrease in circulating leptin concentrations [117]. Therefore, alterations in ghrelin levels as a result of changes in body fat mass may therefore be secondary to changes in leptin [117]. In addition, insulin may also mediate some of the effects J. Jürimäe of body adiposity on circulating ghrelin [117] as circulating ghrelin concentrations are inversely correlated with insulin and insulin resistance values [123]. It has been suggested that relatively low ghrelin concentrations observed in obese individuals may be a result of insulin resistance that is a characteristic in obesity and which has an inhibitory effect on ghrelin concentrations, rather than excess body mass by itself [123]. Collectively, this may represent one mechanism by which insulin is implicated in the homeostatic regulation of energy balance [117]. Only few studies have investigated ghrelin response to different exercise training periods in adult male [30, 114–116] and female [29] athletes. Specifically, in male athletes, ghrelin responses to a weight reduction period before competitions in bodybuilders [30], an intensive training camp in football players [116] and a high-volume lowintensity endurance [114] and a high-volume lowintensity concurrent endurance and resistance [115] training periods in competitive rowers have been studied. In addition, ghrelin responses to intensified training period were also studied in female synchronized swimmers [29]. While studies with national-level male bodybuilders [30] and international-level female synchronized swimmers [29] demonstrated that total ghrelin levels increased together with a body weight reduction as a result of negative energy balance, no differences in total ghrelin concentrations together with no changes in body mass values were observed in competitive male rowers as a result of increased training volume [114, 115]. Accordingly, it can be speculated that body weight loss is also important to reduce total ghrelin concentrations in studied athletes. In contrast, circulating acylated ghrelin concentrations were significantly lowered during the 9-day intensive training camp, which tripled the training volume in male college-level footballers [116]. Therefore, no changes in body mass values were observed, and an increase in physiological stress was associated with a decrease in appetite [116]. It was suggested that an earlyphase physiological stress response may decrease the acylated ghrelin concentrations in male athletes during an intensive training camp [116]. The reason for different results between this study with 12 Ghrelin Responses to Acute Exercise and Training other studies in athletes is not clear. It is possible that these discrepancies are due to factors related to the different modes of exercise, energy availability and competitive level of athletes. However, it is also likely that differences in dietary control, sample collection and assay procedures may also be implicated [117]. Clearly, further studies with elite athletes with different training programmes are needed before any definitive conclusions can be drawn. Relative to women athletes, a national team of female synchronized swimmers performed a 4-week intensified training period, where a baseline training load of about 22 h was increased by a 20.5% across the intensified training period, which caused a significant decrease in body fat percent from 17.3% to 16.4% in these elite female athletes [29]. In addition, a decrease in energy availability was observed, which was accompanied by an increase in ghrelin and decrease in leptin, reflecting a decrease in energy stores across the investigation period [29]. The results of the Schaal et al.’s [29] study demonstrate that a state of an increased fatigue and rather low energy availability in these elite female athletes was characterized by a significant increase in ghrelin levels shortly before the season’s target competitions. Accordingly, it may be suggested that an increased ghrelin concentrations can be used as a marker of increased training stress and inadequate energy availability in elite female athletes. In a study with male bodybuilders, 14 athletes were divided into seven competitors and seven control athletes, who were followed for 11 weeks before the national championships [30]. Competitors were able to significantly decrease their mean body mass by 4.1 kg during the 11-week period, whereas no changes in body composition or ghrelin values were observed in the control athletes [30]. In competitors’ group, the energy deficit at about 536 kcal/day after the first 5-week period was already sufficient to cause a significant increase in total ghrelin concentrations, whereas no further increase in ghrelin levels was observed with the energy deficit reaching 978 kcal/day after 11-week preparatory period [30]. The athletes in the present investigation were competitive bodybuilders with a mean 201 body fat percent of 9.6% at the beginning of the study and 6.5% at the end of the study [30]. It was argued that ghrelin secretion might have reached its limits at some point, and the negative energy balance of more than 900 kcal/day and a significant body weight loss of 2.4 kg in the second 5-week training period (between weeks 6 and 11) were not sufficient to further the significant total ghrelin increase in these athletes [30]. It was concluded that circulating ghrelin levels increase in well-trained bodybuilders with relatively low body fat percent but reach a plateau beyond which there is no further increase in total ghrelin levels, despite continuing negative energy balance and body weight loss [30]. In studies with male rowers, total ghrelin concentrations were measured after a reference week with usual training volume, after 2 weeks of high-volume training and after a recovery week with reduced training volume [114, 115]. In the first study, 90% of the trainings (rowing, running or cycling) were aerobic type of exercise and only 10% resistance type of exercise [114], while in the second study about 50% of the trainings were low-intensity resistance exercise and 50% aerobic type of exercise [115]. It appeared that fasting ghrelin concentrations were not increased as a result of the 2-week period of extended training volume in both studies [114, 115], while a decrease in fasting ghrelin was observed after a recovery week [115]. Although energy intake and energy expenditure increased significantly, the negative energy balance after the 2-week period of high-volume training and energy restriction was about 455 and 408 kcal/day in endurance [114] and concurrent resistance and endurance [115] training studies, respectively. It could be argued that during specific metabolic conditions resulting from the preceding high-volume training period with high energy expenditure, negative energy balance, temporarily restricted caloric condition in fasting state and probably relatively low body energy stores (i.e. low body fat percent) may all contribute to further exercise-induced effects on energy expenditure that leads to ­downregulation of ghrelin concentration in male rowers [114, 115]. 202 Conclusions and Future Directions Energy homeostasis is regulated by a neuroendocrine system that also includes different appetite hormones including ghrelin. Ghrelin concentrations decrease during growth and pubertal maturation and are linked to nutritional status, with lower levels in obese and higher levels in underweight individuals. Therefore, basal ghrelin levels are elevated in growing athletes, while pubertal onset decreases ghrelin levels even in the presence of chronically elevated energy expenditure in young athletes. Since increased participation of children in competitive sport is evident, more research on the exercise-induced modification of the appetite hormones including ghrelin is warranted. It has to be considered that in those sport disciplines where heavy training with large energy expenditure starts at a relatively young age, there is a greater risk for developing the female athletic triad already during adolescent period. It can be suggested that growing and maturing athletes should be monitored at short intervals to better understand the influence of high athletic activity on hormonal markers including ghrelin that are involved in overall growth and energy homeostasis. Ghrelin can be used as an indicator of energy imbalance across the menstrual cycle in female athletes. Elevated ghrelin concentrations have been observed in female athletes with chronic energy deficiency, and ghrelin may differentiate between athletes who will or will not develop functional hypothalamic amenorrhea and be at risk for Relative Energy Deficiency Syndrome in Sport (RED-S). In addition, most of the investigations have studied the role of ghrelin in energy availability in different groups of obese individuals, while less studies have been done with athletes to investigate the possibility to use circulating ghrelin as a possible marker of training stress. The current available information regarding the role of different forms of ghrelin concentrations in energy balance during acute exercise and prolonged training stress is not entirely clear. Acute exercise studies have demonstrated varied responses of different ghrelin forms to the acute exercise in individuals with different body compo- J. Jürimäe sition and physical activity levels. Various investigations with healthy untrained individuals and also well-trained endurance athletes would suggest that exercise-induced acute negative energy balance may not be sufficient to alter total ghrelin and/or acylated ghrelin response. There are also studies that have argued that a certain threshold reduction in energy availability should be reached before any significant postexercise increases in total and/or acylated ghrelin levels occur and that the amplitude of the ghrelin increase could be linked to the energetic status induced by acute exercise stress and the resting levels of ghrelin in athletes. However, to what extent exercise intensity may influence circulating ghrelin response to acute exercise has not yet exactly been determined, although it has been suggested that low- rather than high-intensity exercise with longer duration stimulates ghrelin response. In contrast, different studies with acylated ghrelin have mostly reported significant suppression in measured acylated ghrelin concentration when performed at higher intensities. Therefore, the transient suppression of circulating acylated ghrelin that can be observed during acute exercise may be entirely unrelated to appetite regulation. The differences in ghrelin responses to acute exercise can be attributed to subject physical fitness, pre-exercise meal consumption and timing, the timing of the hormone measurements as well as sampling processing and assay protocols. Additional research is needed to elucidate the exact mechanisms regulating ghrelin synthesis and clearance during and after acute exercise. Results regarding the influence of exercise training programme on circulating ghrelin are more consistent and mainly suggest that exercise training per se has no impact on circulating ghrelin levels, and changes in ghrelin concentrations as a result of exercise training intervention take place as secondary to body weight loss. Therefore, the majority of training studies have investigated the responses of total ghrelin concentrations, with relatively less studies measuring acylated ghrelin separately. Typically, circulating ghrelin levels increase with body weight loss and decrease with body weight gain. Data on ghrelin responses to prolonged exercise training are 12 Ghrelin Responses to Acute Exercise and Training mainly available from obese individuals, whereas only limited data are provided for athletes. It has been suggested that there is a negative energy balance and/or body weight loss threshold to increase circulating ghrelin concentrations that has not yet been exactly determined. It appears that basal and postexercise ghrelin responses without altering body mass are not sensitive enough to represent changes in training volume and energy availability in athletes. There is also some evidence to suggest that although ghrelin increases together with body weight loss in highly trained athletes with already relatively low body fat mass, there may be a plateau beyond which there is no further increase in circulating ghrelin concentrations despite continuing negative energy balance and body weight loss. Further investigations are needed to describe the exact role of ghrelin at different training conditions in athletes representing different sport events. Collectively, additional research including longitudinal studies in different populations with various body composition and physical activity patterns is warranted to better describe the role of ghrelin and its specific forms in conditions of energy deficiency, surplus and balance. References 1. Jürimäe J, Mäestu J, Jürimäe T, et al. Peripheral signals of energy homeostasis as possible markers of training stress in athletes: a review. Metabolism. 2011;60:335–50. 2. Jürimäe J. Adipocytokine and ghrelin responses to acute exercise and sport training in children during growth and maturation. Pediatr Exerc Sci. 2014;26:392–403. 3. Schwartz MW, Woods SC, Porte D Jr, et al. Central nervous system control of food intake. Nature. 2000;404:661–71. 4. Cheng HL, Sainsbury A, Garden F, et al. Ghrelin and peptide YY during puberty: relationships with adolescent growth, development, and obesity. J Clin Endocrinol Metab. 2018;103:2851–60. 5. Suzuki K, Jayasena CN, Bloom SR. The gut hormones in appetite regulation. J Obes. 2011;2011:528401. 6. King JA, Wasse LK, Stensel DJ. The acute effect of swimming on appetite, food intake and plasma acylated ghrelin. J Obes. 2011;2011:351628. 7. Wren AM, Seal LJ, Cohen MA, et al. Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab. 2001;86:4992–5. 203 8. Kojima M, Hosoda H, Date Y, et al. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402:656–60. 9. Fugua IS, Rogol AD. Neuroendocrine alterations in the exercising human: implications for energy homeostasis. Metabolism. 2013;62:911–21. 10. Cummings DE. Ghrelin and the short- and long-­ term regulation of appetite and body weight. Physiol Behav. 2006;30:71–84. 11. Murphy KG, Bloom SR. Gut hormones and the regulation of energy homeostasis. Nature. 2006;444:854–9. 12. Scheid JL, De Souza MJ. Menstrual irregularities and energy deficiency in physically active women: the role of ghrelin, peptide YY and adipocytokines. Med Sport Sci. 2010;55:82–102. 13. Leidy HJ, Dougherty KA, Frye BR, et al. Twenty-­ four-­hour ghrelin is elevated after caloric restriction and exercise training in non-obese women. Obesity. 2007;15:446–55. 14. St-Pierre DH, Faraj M, Karelis AD, et al. Lifestyle behaviours and components of energy balance as independent predictors of ghrelin and adiponectin in young non-obese women. Diabetes Metab. 2006;32:131–9. 15. Leidy HJ, Williams NI. Meal energy content is related to features of meal-related ghrelin profiles across a typical day of eating in non-obese premenopausal women. Horm Metab Res. 2006;38:317–22. 16. Casazza K, Hanks LJ, Alvarez JA. Role of various cytokines and growth factors in pubertal development. Med Sport Sci. 2010;55:14–31. 17. Kraemer RR, Castracane VD. Exercise and humoral mediators of peripheral energy balance: ghrelin and adiponectin. Exp Biol Med. 2007;232:184–94. 18. Muccioli G, Tschop M, Papotti M, et al. Neuroendocrine and peripheral activities of ghrelin: implications in metabolism and obesity. Eur J Pharmacol. 2002;440:235–54. 19. Schwarz NA, Rigby BR, La Bounty P, et al. A review of weight control strategies and their effects on the regulation of hormonal balance. J Nutr Metab. 2011;2011:237932. 20. Asakawa A, Inui A, Fujimiya M, et al. Stomach regulates energy balance via acylated ghrelin and desacyl ghrelin. Gut. 2005;54:18–24. 21. Soares JB, Leite-Moreira AF. Ghrelin, des-acyl ghrelin and obestatin: three pieces of the same puzzle. Peptides. 2008;29:1255–70. 22. Blijdorp K, van der Lely AJ, van den Heuvel-Eibrink MM, et al. Desacyl ghrelin is influenced by changes in insulin concentration during an insulin tolerance test. Growth Hormon IGF Res. 2013;23:193–5. 23. Cederberg H, Rajala U, Koivisto VM, et al. Unacylated ghrelin is associated with changes in body composition and body fat distribution during long-term exercise intervention. Eur J Endocrinol. 2011;165:243–8. 24. Cederberg H, Koivisto VM, Jokelainen J, et al. Unacylated ghrelin is associated with changes in J. Jürimäe 204 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. insulin sensitivity and lipid profile during an exercise intervention. Clin Endocrinol. 2012;76:39–45. Akimizu T, Shinomiya T, Irako T, et al. Separate measurement of plasma levels of acylated and desacyl ghrelin in healthy subjects using a new direct ELISA assay. J Clin Endocrinol Metab. 2005;90:6–9. Lucidi P, Murdolo G, Di Loreto C, et al. Meal intake similarly reduces circulating concentrations of octanoyl and total ghrelin in humans. J Endocrinol Investig. 2004;27:RC12–5. Marzullo P, Verti B, Savia G, et al. The relationship between active ghrelin and human obesity involves alterations in resting energy expenditure. J Clin Endocrinol Metab. 2004;89:936–9. Mackelvie KJ, Meneilly GS, Elahi D, et al. Regulation of appetite in lean and obese adolescents after exercise: role of acylated and desacyl ghrelin. J Clin Endocrinol Metab. 2007;92:648–54. Schaal K, Tiollier E, Le Meur Y, et al. Elite synchronized swimmers display decreased energy availability during intensified training. Scand J Med Sci Sports. 2017;27:925–34. Mäestu J, Jürimäe J, Valter I, et al. Increases in ghrelin and decreases in leptin without altering adiponectin during extreme weight loss in male competitive bodybuilders. Metabolism. 2008;57:221–5. Whatmore AJ, Hall CM, Jones J, et al. Ghrelin concentrations in healthy children and adolescents. Clin Endocrinol. 2003;59:649–54. Pomerants T, Tillmann V, Jürimäe J, et al. Relationship between ghrelin and anthropometrical, body composition parameters and testosterone levels in boys at different stages of puberty. J Endocrinol Investig. 2006;29:962–7. Budak E, Fernandez-Sanchez SM, Believer J, et al. Interactions of the hormones leptin, ghrelin, adiponectin, resistin, and PYY3-36 with the reproductive system. Fertil Steril. 2006;85:1563–81. Jürimäe J, Cicchella A, Jürimäe T, et al. Regular physical activity influences plasma ghrelin concentration in adolescent girls. Med Sci Sports Exerc. 2007;39:1736–41. Võsoberg K, Tillmann V, Tamm AL, et al. Adipocytokine and ghrelin levels in relation to body composition in rhythmic gymnasts entering into puberty: a three-year follow-up study. Pediatr Exerc Sci. 2014;26:477–84. Jürimäe J. Hormones and training. In: Armstrong N, van Mechelen W, editors. Oxford textbook of children’s sport and exercise medicine. 3rd ed. Oxford: Oxford University Press; 2017. p. 455–64. Pomerants T, Tillmann V, Karelson K, et al. Ghrelin response to acute aerobic exercise in boys at different stages of puberty. Horm Metab Res. 2006;38:752–7. Remmel L, Tillmann V, Purge P, et al. Associations of serum leptin, ghrelin and peptide YY levels with physical activity and cardiorespiratory fitness in adolescent boys with different BMI values. Biol Sport. 2017;34:345–52. 39. Parm AL, Jürimäe J, Saar M, et al. Bone mineralization in rhythmic gymnasts before puberty: no longitudinal associations with adipocytokine and ghrelin levels. Horm Res Paediatr. 2012;77:369–75. 40. Jürimäe J, Lätt E, Haljaste K, et al. A longitudinal assessment of ghrelin and bone mineral density with advancing pubertal maturation in adolescent female athletes. J Sports Med Phys Fitness. 2010;50:343–9. 41. Jürimäe J, Lätt E, Haljaste K, et al. Influence of puberty on ghrelin and BMD in athletes. Int J Sports Med. 2009;30:403–7. 42. Kraemer RR, Castracane VD. Effect of acute and chronic exercise on ghrelin and adipocytokines during pubertal development. Med Sport Sci. 2010;55:156–73. 43. St-Pierre DH, Karels AD, Cianflone K, et al. Relationship between ghrelin and energy expenditure in healthy young women. J Clin Endocrinol Metab. 2004;89:5993–7. 44. Parm AL, Jürimäe J, Saar M, et al. Plasma adipocytokine and ghrelin levels in relation to bone mineral density in prepubertal rhythmic gymnasts. J Bone Miner Metab. 2011;29:717–24. 45. Ariyasu H, Takaya K, Tagami T, et al. Stomach is a major source of circulating ghrelin, and feeding state determines plasma ghrelin-like immunoreactivity levels in humans. J Clin Endocrinol Metab. 2001;86:4753–8. 46. Hansen TK, Dall R, Hosoda H, et al. Weight loss increases circulating levels of ghrelin in human obesity. Clin Endocrinol. 2002;56:203–6. 47. Tschop M, Weyer C, Tataranni PA, et al. Circulating ghrelin levels are decreased in human obesity. Diabetes. 2001;50:707–9. 48. Christo K, Cord J, Mendes N, et al. Acylated ghrelin and leptin in adolescent athletes with amenorrhea, eumenorrheic athletes and controls: a cross-sectional study. Clin Endocrinol. 2008;69:628–33. 49. Misra M, Miller KK, Kuo K, et al. Secretory dynamics of ghrelin in adolescent girls with anorexia nervosa and healthy adolescents. Am J Physiol Endocrinol Metab. 2005;289:E373–81. 50. King JA, Wasse LK, Broom DR, et al. Influence of brisk walking on appetite, energy intake, and plasma acylated ghrelin. Med Sci Sports Exerc. 2010;42:485–92. 51. Ravussin E, Tschop M, Morales S, et al. Plasma ghrelin concentration and energy balance: overfeeding and negative energy balance studies in twins. J Clin Endocrinol Metab. 2001;86:4547–51. 52. Gueugnon C, Mougin F, Nguyen NU, et al. Ghrelin and PYY levels in adolescents with severe obesity: effects of weight loss induced by long-term exercise training and modified food habits. Eur J Appl Physiol. 2012;112:1797–805. 53. Choi KM, Lee J, Lee KW, et al. The associations between plasma adiponectin, ghrelin levels and cardiovascular risk factors. Eur J Endocrinol. 2004;150:715–8. 12 Ghrelin Responses to Acute Exercise and Training 54. Kelishadi R, Hashemipour M, Mohammadifard N, et al. Short- and long-term relationships of serum ghrelin with changes in body composition and the metabolic syndrome in prepubescent obese children following two different weight loss programme. Clin Endocrinol. 2008;69:721–9. 55. Jürimäe J, Cicchella A, Tillmann V, et al. Effect of pubertal development and physical activity on ghrelin concentration in boys. J Endocrinol Investig. 2009;32:18–22. 56. Jürimäe J, Kums T, Jürimäe T. Plasma ghrelin concentration is a signal of decreased fat free mass in healthy elderly women. Am J Hum Biol. 2009;21:404–6. 57. Bunt JC, Salbe AD, Tschop MH, et al. Cross-­ sectional and prospective relationships of fasting plasma ghrelin concentrations with anthropometric measures in Pima Indian children. J Clin Endocrinol Metab. 2003;88:3756–61. 58. Park HS, Lee KU, Kim YS, et al. Relationships between fasting plasma ghrelin levels and metabolic parameters in children and adolescents. Metabolism. 2005;54:925–9. 59. Lean MEJ, Malkova D. Altered gut and adipose tissue hormones in overweight and obese individuals: cause or consequence? Int J Obes. 2016;40:622–32. 60. Ata SM, Vaishnav U, Puglisi M, et al. Macronutrient composition and increased physical activity modulate plasma adipokines and appetite hormones during a weight loss intervention. J Women Health. 2010;19:139–45. 61. Cummings DE, Weigle DS, Frayo RS, et al. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346:1623–30. 62. Foster-Schubert KE, McTiernan A, Scott Frayo R, et al. Human plasma ghrelin levels increase during a one-year exercise program. J Clin Endocrinol Metab. 2005;90:820–5. 63. Hagobian TA, Sharoff CG, Stephens BR, et al. Effects of exercise on energy-regulating hormones and appetite in men and women. Am J Physiol Regul Integr Comp Physiol. 2009;296:R233–42. 64. Scheid JL, De Souza MJ, Leidy HJ, et al. Ghrelin but not peptide YY is related to change in body weight and energy availability. Med Sci Sports Exerc. 2011;43:2063–71. 65. Guelfi KJ, Donges CE, Duffield R. Beneficial effects of 12 weeks of aerobic compared with resistance exercise training on perceived appetite in previously sedentary overweight and obese men. Metabolism. 2013;62:235–43. 66. King JA, Wasse LK, Ewens J, et al. Differential acylated ghrelin, peptide YY3-36, appetite, and food intake responses to equivalent energy deficits created by exercise and food restriction. J Clin Endocrinol Metab. 2011;96:1114–21. 67. Burns SF, Broom DR, Miyashita M, et al. A single session of treadmill running has no effect on plasma total ghrelin concentrations. J Sports Sci. 2007;25:635–42. 205 68. Dall R, Kanaley J, Hansen TK, et al. Plasma ghrelin levels during exercise in healthy subjects and in growth hormone-deficit patients. Eur J Endocrinol. 2002;147:65–70. 69. Christ ER, Zehnder M, Boesch C, et al. The effect of increased lipid intake on hormonal responses during aerobic exercise in endurance-trained men. Eur J Endocrinol. 2006;154:397–403. 70. Erdmann J, Tahbaz R, Lippl F, et al. Plasma ghrelin levels during exercise – effects of intensity and duration. Regul Pept. 2007;143:127–35. 71. Jürimäe J, Hofmann P, Jürimäe T, et al. Plasma ghrelin responses to acute sculling exercises in elite male rowers. Eur J Appl Physiol. 2007;99:467–74. 72. Jürimäe J, Jürimäe T, Purge P. Plasma ghrelin is altered after maximal exercise in elite male rowers. Exp Biol Med. 2007;232:904–9. 73. Jürimäe J, Rämson R, Mäestu J, et al. Plasma visfatin and ghrelin response to prolonged sculling in competitive male rowers. Med Sci Sports Exerc. 2009;41:137–43. 74. Kraemer RR, Durand RJ, Acevedo EO, et al. Rigorous running increases growth hormone and insulin-like growth factor-I without altering ghrelin. Exp Biol Med. 2004;229:240–6. 75. Kraemer RR, Durand RJ, Hollander DB, et al. Ghrelin and other glucoregulatory hormone responses to eccentric and concentric muscle contractions. Endocrine. 2004;24:93–8. 76. Malkova D, McLaughlin R, Manthou E, et al. Effect of moderate-intensity exercise session on preprandial and postprandial responses of circulating ghrelin and appetite. Horm Metab Res. 2008;40:410–5. 77. Toshinai K, Kawagoe T, Shimbara T, et al. Acute incremental exercise decreases plasma ghrelin levels in healthy men. Horm Metab Res. 2007;39:849–51. 78. Schmidt A, Maier C, Schaller G, et al. Acute exercise has no effect on ghrelin plasma concentrations. Horm Metab Res. 2004;36:174–7. 79. Borer KT, Wuorinen E, Chao C, et al. Exercise energy expenditure is not consciously detected due to oro-gastric, not metabolic, basis of hunger sensation. Appetite. 2005;45:177–81. 80. Ghanabar-Niaki A. Ghrelin and glucoregulatory hormone responses to a single circuit resistance exercise in male collegiate students. Clin Biochem. 2006;39:966–70. 81. Vestergaard ET, Dall R, Lange KHW, et al. The ghrelin response to exercise before and after growth hormone administration. J Clin Endocrinol Metab. 2007;92:297–303. 82. Sartorio A, Morpurgo P, Capiello V, et al. Exercise-­ induced effects on growth hormone levels are ­associated with ghrelin changes only in the presence of prolonged exercise bouts in male athletes. J Sports Med Phys Fitness. 2008;48:97–101. 83. Broom DR, Stenzel DJ, Bishop NC, et al. Exercise-­ induced suppression of acylated ghrelin in humans. J Appl Physiol. 2007;102:2165–71. 206 84. Broom DR, Miyashita M, Wasse LM, et al. Acute effect of exercise intensity and duration on acylated ghrelin and hunger in men. J Endocrinol. 2017;232:411–22. 85. King JA, Miyashita M, Wasse LK, et al. Influence of prolonged treadmill running on appetite, energy intake and circulating concentrations of acylated ghrelin. Appetite. 2010;54:492–8. 86. Shiiya T, Ueno H, Toshinai K, et al. Significant lowering of plasma ghrelin but not des-acyl ghrelin in response to acute exercise in men. Endocr J. 2011;58:335–42. 87. Deighton K, Stensel DJ. Creating an acute energy deficit without stimulating compensatory increases in appetite: is there an optimal exercise protocol? Proc Nutr Soc. 2014;73:352–8. 88. Schubert MM, Desbrow B, Sabathy S, et al. Acute exercise and subsequent energy intake. A meta-­ analysis. Appetite. 2013;63:92–104. 89. Holliday A, Blannin A. Appetite, food intake and gut hormone responses to intense aerobic exercise of different duration. J Endocrinol. 2017;235:193–205. 90. Kojima C, Ishibashi A, Ebi K, et al. The effect of a 20 km run on appetite regulation in long distance runners. Nutrients. 2016;8:672. 91. Larson-Meyer DE, Palm S, Bansal A, et al. Influence of running and walking on hormonal regulators of appetite in women. J Obes. 2012;2012:7304409. 92. Mattin LR, Yau AMW, McIver V, et al. The effect of exercise intensity on gastric emptying rate, appetite and gut derived hormone responses after consuming a standardized semi-solid meal in healthy males. Nutrients. 2018;10:787. 93. Sauseng W, Nagel B, Gamillscheg A, et al. Acylated ghrelin increases after controlled short-time exercise in school-aged children. Scand J Med Sci Sports. 2011;21:e100–5. 94. Sim AK, Wallman KE, Fairchild TJ, et al. High-­ intensity intermittent exercise attenuates ad-libitum energy intake. Int J Obes. 2014;38:417–22. 95. Howe SM, Hand TM, Larson-Meyer DE, et al. No effect of exercise intensity on appetite in highly-­ trained endurance women. Nutrients. 2016;8:223. 96. Wasse LK, Sunderland C, King JA, et al. The influence of vigorous running and cycling exercise on hunger perceptions and plasma acylated ghrelin concentrations in lean young men. Appl Physiol Nutr Metab. 2013;38:1–6. 97. Becker GF, Macedo RC, Cunha Gdos S, et al. Combined effects of aerobic exercise and high-­ carbohydrate meal on plasma acylated ghrelin and levels of hunger. Appl Physiol Nutr Metab. 2012;37:184–92. 98. Goltz FR, Thackray AE, King JA, et al. Interindividual responses of appetite to acute exercise: a replicated crossover study. Med Sci Sports Exerc. 2018;50:758–68. 99. Schubert MM, Sabapathy S, Leveritt M, et al. Acute exercise and hormones related to appetite regulation: a meta-analysis. Sports Med. 2014;44:387–403. J. Jürimäe 100. Smith JK. Exercise, obesity and CNS control of metabolic homeostasis: a review. Front Physiol. 2018;9:574. 101. Douglas JA, King JA, McFarlane E, et al. Appetite, appetite hormone and energy intake responses to two consecutive days of aerobic exercise in healthy young men. Appetite. 2015;92:57–65. 102. Russel RR, Willis KS, Ravussin E, et al. Effect of endurance running and dietary fat on circulating ghrelin and PYY: potential role in appetite regulation. J Sports Sci Med. 2009;8:574–83. 103. Hazell TJ, Islam H, Townsend LK, et al. Effects of exercise intensity on plasma concentrations of appetite-­ regulating hormones: potential mechanisms. Appetite. 2016;98:80–8. 104. Garcia JM, Iyer D, Poston WS, et al. Rise of plasma ghrelin with weight loss is not sustained during weight maintenance. Obesity. 2006;14:1716–23. 105. Flack KD, Ufholz K, Johnson LA, et al. Energy compensation in response to aerobic exercise training in overweight adults. Am J Physiol Regul Integr Comp Physiol. 2018;315:R619–26. 106. Kang SJ, Kim JH, Gang Z, et al. Effects of 12-week circuit exercise program on obesity index, appetite regulating hormones, and insulin resistance in middle-aged obese females. J Phys Ther Sci. 2018;30:169–73. 107. Mason C, Xiao L, Imayama I, et al. The effects of separate and combined dietary weight loss and exercise on fasting ghrelin concentrations in overweight and obese women: a randomized controlled trial. Clin Endocrinol. 2015;82:369–76. 108. Leidy HJ, Gardner JK, Frye BR, et al. Circulating ghrelin is sensitive to changes in body weight during a diet and exercise program in normal weight young women. J Clin Endocrinol Metab. 2004;89:2659–64. 109. Morpurgo PS, Resnik M, Agosti F, et al. Ghrelin secretion in severely obese subjects before and after a 3-week integrated body mass reduction program. J Endocrinol Investig. 2003;26:723–7. 110. Gibbons C, Blundell JE, Caudwell P, et al. The role of episodic postprandial peptides in exercise-­ induced compensatory eating. J Clin Endocrinol Metab. 2017;102:4051–9. 111. Bowyer KP, Carson JA, Davis JM, et al. The influence of exercise training dose on fasting acylated ghrelin concentration in older women. J Behav Med. 2019;42(3):567–72. 112. Otto B, Cuntz U, Fruehauf E, et al. Weight gain decreases elevated plasma ghrelin concentrations in patients with anorexia nervosa. Eur J Endocrinol. 2001;154:669–73. 113. Martins C, Kulseng B, King NA, et al. The effects of exercise-induced weight loss on appetite-related peptides and motivation to eat. J Clin Endocrinol Metab. 2010;95:1609–16. 114. Rämson R, Jürimäe J, Jürimäe T, et al. The influence of increased training volume on cytokines and ghrelin concentration in college level male rowers. Eur J Appl Physiol. 2008;104:839–46. 12 Ghrelin Responses to Acute Exercise and Training 115. Rämson R, Jürimäe J, Jürimäe T, et al. The effect of 4-week training period on plasma neuropeptide Y, leptin and ghrelin responses in male rowers. Eur J Appl Physiol. 2012;112:1873–80. 116. Oshima S, Takahato C, Sasahara I, et al. Changes in stress and appetite responses in male power-trained athletes during intensive training camp. Nutrients. 2017;9:912. 117. King JA, Wasse LK, Stensel DJ, et al. Exercise and ghrelin. A narrative overview of research. Appetite. 2013;68:83–91. 118. Russell M, Misra M. Influence of ghrelin and adipocytokines on bone mineral density in adolescent female athletes with amenorrhea and eumenorrheic athletes. Med Sport Sci. 2010;55:103–13. 119. Maimoun L, Georgopoulos NA, Sultan C. Endocrine disorders in adolescent and young female athletes: impact on growth, menstrual cycles, and bone mass acquisition. J Clin Endocrinol Metab. 2014;99:4037–50. 207 120. Ackerman KE, Slusarz KM, Guereca H, et al. Higher ghrelin and lower leptin secretion are associated with lower LH secretion in young amenorrheic athletes compared with eumenorrheic athletes and controls. Am J Physiol Endocrinol Metab. 2012;302:E800–6. 121. De Souza MJ, Lee DK, Van Heest JL, et al. Severity of energy-related menstrual disturbances increases in proportion to indices of energy conservation in exercising women. Fertil Steril. 2007;88:971–5. 122. Kasa-Vubu JZ, Rosenthal A, Murdock EG, et al. Impact of fatness, fitness, and ethnicity on the relationship of nocturnal ghrelin to 24-hour luteinizing hormone concentrations in adolescent girls. J Clin Endocrinol Metab. 2007;92:3246–52. 123. King NA, Gibbons CHE, Martins C. Ghrelin and obestatin concentrations during puberty: relationships with adiposity, nutrition and physical activity. Med Sport Sci. 2010;55:69–81. Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise 13 Charles E. Wade Introduction In response to exercise, there are numerous alterations in fluid and electrolyte homeostasis. These perturbations occur immediately upon initiation of exercise and can persist for hours or even days after completion of exercise. The endocrine system plays an important role in the regulation of fluid and electrolyte homeostasis that must occur with exercise. Dysregulation of the endocrine system may limit exercise activity and, in some incidences, result in debilitating morbidities or death. This chapter emphasizes responses to exercise and reviews the importance and factors involved in the maintenance of fluid and electrolyte balance. Previous reviews will be used to address the basics of effected systems; however, emphasis is placed on new data and the current discussions about performance of work and exercise. The term exercise is an ambiguous term covering a broad range of physical activities. The term is employed to define activities such as running and cycling but is also used to cover the activities of daily living and work. Thus, when discussing responses to exercise, it is important to clarify the type of activity, the level at which it C. E. Wade (*) Center for Translational Injury Research (CeTIR), Houston, TX, USA e-mail: charles.e.wade@uth.tmc.edu is performed, and the duration. In defining the responses to exercise, it is essential to understand the definitions of workload. The absolute workload is the level of exercise being performed, such as running on a treadmill at a defined speed. For individual subjects, this would produce a variable response depending on their level of fitness/training. Therefore, to compare exercise responses between subjects, relative workload is often employed as a normalization technique [1–3]. Relative workload is expressed as a percentage of the maximum capability of the individual to perform that specific exercise and is often further standardized to the heart rate or oxygen consumption of the subject. Physiologic Responses to Exercise A variety of conditions results from alterations in fluid and electrolytes and affects the performance of exercise and work. The disruption of the balance of fluids and electrolytes correlates with limitation of work capacity; however, the range of changes tolerated may be extended with training and repeated exposures. In general the body can undergo one of several responses to exercise: dehydration, dysnatremia, hypovolemia, or hypervolemia. The following text will review each. Dehydration is defined as a reduction in total body water (TBW) and an increase in plasma © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_13 209 C. E. Wade 210 electrolyte concentrations. Heavy exercise and extreme heat are two of the most prevalent causes of dehydration, as both are associated with exercise and subsequent loss of fluid volume due to sweating and inadequate fluid intake [4–7]. Current evidence suggest that dehydration resulting in a decrease of greater than 2% body mass will adversely affect exercise performance [6–8]. However, decrements in selfpaced exercise may not occur until a 4% loss in body weight [9]. Regardless of the level of dehydration, a loss of TBW and an increase in plasma electrolyte concentrations are associated with limited work performance and, in extreme cases, death. Dysnatremia covers the occurrence of both increases and decreases in plasma sodium observed with exercise [10–14]. Siegel et al. noted an incidence rate of dysnatremia of 32.5% in 1319 collapsed marathon runners [12]. Of these, 85% were hypernatremic and 15% hyponatremic. Both of these conditions have been associated with deaths in competitive runners. Hypovolemia is a decrease in blood volume in the absence of changes in plasma electrolyte concentrations. This can occur with exercise or hemorrhage [15] and follows periods of water submersion to the neck or the administration of diuretics commonly used in the treatment of hypertension [16, 17]. Hypovolemia necessitates an increase in heart rate at submaximal workloads and a more rapid increase in body temperature, both indicative of limited work performance [7]. In contrast, hypervolemia is the expansion of blood volume. There is extensive literature on the expansion of blood volume by increasing the red cell mass; however, within the scope of this chapter, this term refers to expansion of the plasma volume. Plasma volume is expanded by exercise training and by acute excessive ingestion of fluids, hyperhydration [7, 18–20]. Warburton et al. reviewed the literature on the effect of acute expansion of plasma volume and found minimal increases in maximum oxygen consumption, but there were negligible changes in exercise endurance [20]. Modulation of Hormones in Responses to Exercise Workload Intensity The response of hormones to exercise is closely related to the amount of relative work performed. There are three basic patterns of hormones during exercise. The first is an increase proportional to the increase in relative workload. For example, with each increase in workload, there is a constant increase in the plasma hormone concentration of atrial natriuretic peptide (ANP) (Fig. 13.1a). The second pattern is a logarithmic/ exponential increase such as that reported for plasma renin activity (PRA) (Fig. 13.1b). With increasing workloads, the level of hormone increases at an exponentially faster rate. The third pattern is related to an onset of an increase at a given threshold; this is observed for vasopressin (Fig. 13.1c). A threshold response for exercise is usually associated with the onset of anaerobic metabolism and a relative workload of about 70%. This has also been associated with the increase in stress-related hormones such as cortisol and adrenocorticotropic hormone (ACTH). These patterns of increased hormone concentrations are consistently observed in studies of acute exercise when the response is expressed relative to the workload of the task performed. Exercise Duration The duration of exercise is also a confounding factor in the response of hormones to exercise. Extended time, rather than intensity, may have a greater influence on the levels of hormones during exercise. This is especially true of hormones involved in the regulation of fluid and electrolyte homeostasis. As exercise progresses, there is an increased metabolic heat necessitating sweating and therefore the loss of water and electrolytes. The increase in aldosterone, which regulates sodium balance, is increased twofold with acute maximal exercise (i.e., running on a treadmill) and returns to baseline levels within an hour. 13 a Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise b Linear 300 211 Logarithmic/Exponential 1500 PRA% Resting ANP% Resting 250 200 150 100 1000 500 50 0 0 0 50 0 100 100 % Workload % Workload c 50 Threshold AVP% Resting 1500 1000 500 0 50 0 100 % Workload Fig. 13.1 The various patterns of response of hormones to exercise. The individual dots represent the response from independent studies of exercise on a cycle ergometer with varying workloads. The variance represents differences in how the exercise was performed, state of hydration of the subjects, and difference in assay techniques. With these confounders the patterns in response to exercise are still present. The linear example is demonstrated by the response of atrial natriuretic peptide (ANP; (a) 11 studies), the logarithmic/exponential increase by plasma renin activity (PRA; (b) 20 studies), and the threshold response by vasopressin (AVP; (c) 23 studies) Extended exercise times elicit similar changes in plasma volume and sodium concentrations, but aldosterone concentration increases three to four times the basal levels and remains elevated for over 24 h [21]. The greater and enduring response to exercise of longer duration is postulated to be due to additional regulators associated with the “stress” of exercise [10, 22]. Of note, hormone concentrations may vary over time with exercise of long duration, such as during a marathon or ultra-endurance events. For example, ANP is increased by a factor of ten during the first 10 km of a marathon but subsequently decreased to levels only fivefold greater than baseline [23]. In addition, the conditions under which recovery is conducted, access to fluids or cool down exercise, are influential in the postexercise responses and need to be clarified [24, 25]. Recently, Hew-­ Butler et al. compared the hormonal responses to maximal exercise with a mean duration of 10–60 min of exercise at a treadmill speed equivalent to 60% of the maximum [26]. With maximal acute exercise, significant increases were reported for vasopressin (5-fold) and aldosterone C. E. Wade 212 (2-fold), while at submaximal effort, only aldosterone was increased (3.3-fold). Thus, both the length of time and intensity of the workload must be considered when studying the regulation and function of hormones in response to exercise. Training The level of training of a subject may influence the hormonal response to exercise [27, 28]. While much of the variance between subjects at absolute workloads may be due to differences in the relative workload being performed, there are still aspects of training that change the response. Individuals undergoing persistent heavy bouts of exercise training may have alterations in resting levels. In subjects doing daily long-distance runs, plasma aldosterone concentrations are elevated compared to controls [29]. However, for the majority of hormones regulating fluid and electrolyte homeostasis, training does not appear to be as an important of a factor as the intensity and duration of exercise in the response of these hormones. Hydration Status The initial hydration status of a subject may influence subsequent responses to exercise. Fluid intake during the performance of exercise is also an influencing factor. Dehydration or hyperhydration alters initial hormone levels; however, the subsequent response to exercise appears independent. Geelen et al. found that following dehydration, ingestion of fluid caused a rapid and pronounced reduction in vasopressin and an increase in norepinephrine that was independent of changes in plasma osmolality and volume [30]. No changes were noted in epinephrine, aldosterone, PRA, or ANP. Additional investigations reported that the greater the volume consumed, the more pronounced the decrease in vasopressin and increase in norepinephrine [24, 31–33]. This suggests an oropharyngeal reflex may be present and mediated by the sympathetic nervous system. Khamnei et al. evaluated the effect of the combination of exercise and postexercise fluid intake on vasopressin [24, 32]. Subjects exercised at 50% of their maximum oxygen uptake for 30 min. Exercise resulted in a 45% increase in vasopressin which was sustained after exercise in the absence of fluid intake. In contrast, when a large volume of fluid was ingested after exercise, control levels of vasopressin were obtained within 3 min. These findings suggest fluid intake may have a profound effect on hormonal responses during exercise, independent of changes in plasma volume and osmolality. Hew-­Butler has put forth the hypothesis that inappropriate increases in vasopressin during prolonged exercise in the presence of adequate fluid intake may be a contributing factor to hyponatremia and subsequent morbidity [10, 22]. This line of research awaits additional well-controlled prospective studies to fully identify underlying mechanisms. Sex Sex of the subject is another factor with demonstrated differences. In women, the phase of the menstrual cycle in which exercise is performed may alter the hormonal responses. Resting aldosterone levels are increased during the mid-luteal phase of the cycle, and the response to exercise is amplified [34]. Further work by Stachenfeld and coworkers has demonstrated the effect of progesterone and estrogen on the levels and responses of hormones that are important in fluid and electrolyte homeostasis [35, 36]. In patients with coronary heart disease, basal levels of vasopressin were elevated in men; however, in responses to a 6 min walk test that increased vasopressin, ANP, norepinephrine, and epinephrine, there were no differences between males and females [37]. Following exercise in well-trained subjects to decrease body mass by 3%, women had a lower PRA and faster recovery of aldosterone and slower recovery of vasopressin compared to men [38]. Overall, there are minimal differences reported between male and females in resting hormone levels, and differences in response to exercise are not fully delineated [39]. 13 Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise 213 Health Status Other Influencing Factors The initial health of the subject is an influencing factor in the hormonal responses to exercise and offers insights to the pathophysiology of various disease processes and in some cases a means of diagnosis and/or rehabilitation. The presence of disease represents a shift in homeostasis that requires alteration in the responsiveness of hormones important in fluid and electrolyte homeostasis. In age-matched subjects, Shim and coworkers reported that subjects with an exaggerated blood pressure response to exercise, which is indicative of a greater risk for hypertension and prevalence of cardiac hypertrophy, had elevated levels of angiotensin II at rest and an augmented increase in response to exercise [40]. However, there were no significant differences in norepinephrine, epinephrine, PRA, or aldosterone at the end of exercise. Kjaer et al. studied patients with congestive heart failure (CHF) and compared them with healthy subjects at 50 and 75% of their maximum workloads on a cycle ergometer [41]. Basal levels of ANP, brain natriuretic peptide (BNP), vasopressin, and PRA were elevated in patients with CHF. In response to exercise, ANP, arginine vasopressin (AVP), norepinephrine, and epinephrine were all increased in both groups. Even though higher absolute levels were observed in subjects with CHF, when expressed as a percent of basal concentrations group, differences were negated. BNP was increased with exercise only in patients with CHF. Coiro et al. assessed the response of vasopressin to exercise to exhaustion on a bicycle ergometer in subjects with diabetes and controls and further segregated the groups as smokers and nonsmokers [42]. Baseline vasopressin concentrations at rest (2.1–2.6 pg/mL) were not different between groups. In all groups, there was a significant increase in vasopressin in response to exercise. While smoking was not identified as a contributing factor, there was a greater increase in vasopressin in subjects with diabetes (12– 13 pg/mL) than controls (7–8 pg/mL). The difference between diabetic and normal subjects could not be attributed to cardiovascular or respiratory responses. Other confounders, such as position of exercise and age, have been identified to influence hormonal responses to exertion. Wolf et al. compared supine and upright exercise on a cycle ergometer at a relative workload of 40–50% for 20 min. With supine exercise, the response of PRA and aldosterone to exercise was increased by 90% and 49%, respectively, in contrast to upright exercise [43]. These differences occurred in the absence of difference between the types of exercise in plasma osmolality or blood pressure. Perrault et al. found ANP concentrations to be increased and vasopressin, PRA, and norepinephrine to be reduced, during supine exercise on a cycle ergometer in comparison to exercise in an upright position [44]. During the performance of a marathon, subjects with a mean age of 47 years had an increase in ANP to 104 pg/mL compared to 43 pg/mL in younger subjects with a mean age of 28 years [23]. In addition, differences in hormone concentrations reported in response to exercise may be in part explained by the differing methods of measurement. The presence of such confounders in the comparison of the hormonal responses to exercise has not been systematically addressed, partially limiting our interpretation of the role of hormones in fluid and electrolyte homeostasis during exercise. Hormone Responses to Exercise The hormones of consequence to fluid and electrolyte balance in exercising humans are those involved in the regulation of thirst and function of the kidneys and sweat glands. The essential hormones are the catecholamines, vasopressin, the renin-angiotensin-aldosterone system, and natriuretic peptides. While these hormones have a variety of functions, the focus of the present review will be on their responses to exercise and impact on fluid and electrolyte homeostasis ­during and following exercise. Circulating levels of these hormones are altered during exercise as a function of changes in secretion, C. E. Wade 214 metabolism, and volume of distribution. The most common measurement of these hormones in association with exercise is the circulating concentrations, which will be the focus of the present effort. Catecholamines Catecholamines, specifically norepinephrine and epinephrine, are derived from increases in sympathetic nervous system activity and the adrenal glands [45, 46]. The kidneys are also suggested as a source of norepinephrine [47]. Levels of circulating catecholamines respond rapidly upon the onset of exercise in order to redistribute blood flow to meet metabolic demands [2, 48, 49]. In response to exercise, there is a progressive increase in circulating norepinephrine levels from 1.3 to 3.0 nmol/L at rest to 12.0 nmol/L following maximal exercise [45, 50–52]. The increase in epinephrine occurs later in the course of exercise and can rise from resting levels of 380–655 pmol/L to concentrations over 3000 pmol/L. The increase in the ratio of norepinephrine to epinephrine demonstrates activation of the sympathetic nervous system and is attributed to active spillover from the muscles during exercise [45, 52–54]. With continued exercise, there is an attenuation of the increase in the ratio of norepinephrine to epinephrine, which is indicative of an increase in the release of epinephrine predominately from the adrenal medulla under the control of hypothalamic mediation in addition to the sympathetic nervous system. Following exercise, plasma levels of catecholamines return to resting levels in a matter of minutes, as they have a short half-life due to degradation and reuptake by the sympathetic nervous system. Recent studies that inhibited the reuptake of norepinephrine have demonstrated an increase in the time necessary to complete work equal to 30 min of exercise at 75% of maximal workload [55, 56]. These studies suggest clearance from the circulation of norepinephrine plays a role in fatigue. Vasopressin AVP is also known as vasopressin or antidiuretic hormone (ADH). It is a neurohypophysial hormone synthesized in the hypothalamus and stored in the posterior pituitary [57, 58]. Vasopressin is a pressor that alters peripheral resistance, but its greatest effect is on the reabsorption of water in the collecting tubules of the kidneys. Secretion of vasopressin is regulated by alterations in plasma osmolality and blood pressure. Circulating concentrations of vasopressin in humans are 1–4 pg/ mL [57, 59–62]. With maximal exercise, vasopressin concentrations of 4–24 pg/mL are reported. Maximum conservation of water by the kidneys is observed at vasopressin levels of 10–20 pg/mL. With progressive increases in exercise, elevation of vasopressin is not observed until 70% of maximum workload is attained, i.e., the anaerobic threshold (Fig. 13.1c). Animal experiments have demonstrated an increase in activation of hypothalamic neurons that is indicative of increased vasopressin content (production) and of performing above the anaerobic threshold [63]. Thus, the response of vasopressin appears to be associated with the onset of anaerobic metabolism, which is also related to increases in “stress hormones” such as cortisol and ACTH. An increase in vasopressin may persist for over 60 min after exercise or longer if access to fluids is restricted. Of note, at low workloads of about 25% of the anaerobic threshold, vasopressin decreases have been reported. A variety of factors have been demonstrated to mediate the increase in vasopressin with exercise, including the increase in osmolality and reduction in intravascular volume; however, the increase in plasma osmolality appears to be the primary mediator (Fig. 13.2) [59, 64, 65]. In subjects exercising at 65% of maximum while running on a treadmill, there was a progressive increase in vasopressin with progressive workloads [66]. In subsequent tests which involved dehydration that decreased body weight by 3 and 5%, resting vasopressin levels were increased in association with the decrease in blood volume; however, in response to exercise, further increases 13 a Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise b AVP 10 Thirst Very, very 7 thirsty 6 8 5 6 Thirst Plasma AVP 215 4 4 3 2 2 1 0 275 285 295 Not thirsty 0 280 Plasma Osmolality (mosmol/kg–1) 285 290 295 Plasma Osmolality (mosmol/kg–1) Fig. 13.2 (a) Levels of vasopressin and (b) subjective assessment of thirst in association to plasma changes in osmolality during moderate exercise. Measurements were from subjects with different levels of fitness, under various levels of hydration. (Redrawn from Merry et al. [27]) in vasopressin were related to the magnitude of the increase in osmolality. Brandenberger et al. evaluated rehydration during exercise giving subjects no fluids, water, or an isotonic solution. Intake of water reduced osmolality but did not alter plasma volume [67]. Consumption of the isotonic solution did not change osmolality but increased plasma volume. Both methods of rehydration decreased the rise in vasopressin levels with exercise, as well as those of PRA and cortisol. Others have reported similar findings [32, 44, 68]. The independence of the increase in osmolality and blood volume, and the regulation of vasopressin in response to exercise, is similar to that reported with dehydration. Coiro and colleagues have demonstrated that the increase in vasopressin during exercise to exhaustion may be attenuated by blockade of 5-HT3 serotonergic receptors and administration of somatostatin, supporting another means of mediating the increase in vasopressin during exercise [69]. Recently, Hew-Butler et al. have questioned the relationship of vasopressin and plasma osmolality during exercise. In subjects participating in an ultramarathon, they observed 3.9-fold increase in plasma vasopressin, no significant change in plasma sodium, and a significant decrease in plasma volume [10, 22]. They also evaluated cyclists during a 109 km race and observed nearly identical changes [70]. In subjects participating in an ultramarathon, they observed a 3.9-fold increase in plasma vasopressin in the absence of a significant change in plasma sodium though plasma volume was significantly decreased. These authors and others hypothesize that under conditions of prolonged exercise, the osmotic regulation of vasopressin is overshadowed by non-osmotic stimuli, of which, the reduction in blood volume plays a minor role [14, 71, 72]. The increase in AVP was associated with elevations in cortisol, oxytocin, and BNP, which underscores the relationship of AVP release with “exercise stress.” Irrespective of the means, vasopressin is elevated by more than fourfold during acute exercise to exhaustion or intense prolonged exercise. Renin-Angiotensin-Aldosterone The renin-angiotensin-aldosterone systems are closely coupled and increased in response to exercise. Renin is released from the kidney in response to sympathetic nerve stimulation, as well as norepinephrine spillover, resulting in C. E. Wade 216 Natriuretic Peptides Peptides demonstrated to elicit a natriuresis have been deemed natriuretic peptides. These include ANP, BNP, urodilatin, and adrenomedullin. 300 Urinary Na+ Excretion (µmol/min) increased plasma concentrations [17, 45, 52, 73– 76]. Renin then converts angiotensinogen to angiotensin I, which is subsequently transformed to angiotensin II in the lung. Angiotensin II promotes the release of aldosterone from the adrenal gland. With exercise, all aspects of this system are increased and play a variety of roles in the regulation of fluid and electrolyte homeostasis [3, 45, 60, 77, 78]. At rest, PRA has levels in the order of 0.15–0.55 ng angiotensin I/mL/h and with maximal exercise increases to levels of 1.11–1.67 ng angiotensin I/mL/h. There is an exponential increase in renin activity with increasing workloads; significant differences are reported at levels of 60–70% of maximum (Fig. 13.1b). The increase in PRA with exercise is positively associated with the increase in angiotensin II. Basal levels of angiotensin II are 15–25 ng/L, with values of 130–160 ng/L achieved with maximal exercise. Aldosterone release is regulated by angiotensin II, as well as ACTH and the plasma levels of sodium and potassium. Aldosterone concentration increases from resting levels of 80–830 pmol/L to concentrations of 250– 3330 pmol/L with maximal exercise. Blockade of the conversion of angiotensin I to angiotensin II does not attenuate the response of aldosterone to maximal exercise, which supports the theory that other pertinent regulatory factors are involved [79, 80]. The elevation of aldosterone may persist for days after exercise, and levels are dependent upon the sodium and water intake [21]. In the postexercise period, the increase in aldosterone may be the product of increased water intake, which reduces the plasma sodium concentration or the persistent elevation of aldosterone—which is due to activation of the ACTH. Irrespective of the cause, the increase in aldosterone due to exercise plays a role in the conservation of sodium in the sweat glands and kidneys (Fig. 13.3). 250 200 150 100 50 0 0 200 400 600 Plasma Aldosterone (pg/mL) Fig. 13.3 Plasma aldosterone concentrations were compared to the urinary excretion of sodium at the end of a 2 h run (closed circles) and following 48 h of recovery with food and water ad libitum (open circles). With exercise, there was an increase in aldosterone, and over the recovery period, there was a decrease. (Adapted from Wade et al. [29])) These peptides appear to participate in the regulation of fluid homeostasis by protecting against volume and pressure overloads. Though these peptides have been extensively studied over the past 30 years in patients with disease such as heart failure, pulmonary hypertension, and chronic renal disease, their response to and role during exercise are not well defined. Additionally, well-designed studies in control subjects or during competitive events have yet to be undertaken. trial Natriuretic Peptide A ANP is increased with exercise in a linear response (Fig. 13.1a). Resting plasma levels of 10–49 pg/mL are increased to over 100 pg/mL with acute maximal exercise [22, 25, 39, 44, 81, 82]. In response to long-duration exercise, there is initially a pronounced increase, a subsequent fall, and then a re-elevation of levels, persisting until completion of the exercise [23]. Resting levels are obtained within hours of cessation of the activity [77]. The primary stimulus for the increase in ANPwith acute exercise is an increase in atrial stretch due to an increase in venous return [62]. However as exercise progresses, atrial pressure decreases as blood flow is 13 Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise redistributed (cardiovascular drift) to meet the metabolic demand of active tissues and to dissipate the thermal load [48, 49]. The response of ANP to extended exercise may be increased if water is ingested, suggesting a fluid volume change directly on the heart mediating release [83, 84]. Recently, pronounced increases in ANP with exercise have been associated with increases in cardiac troponin levels, suggesting myocardial damage during heavy exercise could be a contributing factor to increases in ANP [85]. In cardiac transplant patients, ANP levels are elevated, and the response to exercise is accentuated. This suggests that in normal subjects with naturally innervated hearts, there may be neural inhibition of ANP release [44, 86, 87]. Support for this hypothesis is the observation in patients with hypertension that chronic beta-blockade substantially increases the ANP response to exercise [88]. Sodium intake appears to also affect the ANP response to exercise [89, 90]. During submaximal cycle ergometer exercise when subjects were on a low-sodium diet, ANP increased from 42 to 59 pg/mL, in contrast to a high-sodium diet where the increase was from 72 to 119 pg/ mL. Thus, the increase in ANP with exercise appears to be related to a number of factors: stretch of the atrium due to volume changes, neurological inputs, and sodium intake. rain Natriuretic Peptide B BNP, as its name implies, was first identified in the brain and subsequently identified in other tissues, specifically in the heart [91, 92]. BNP is collocated with ANP in the heart and appears to have similar paths of regulation and actions. BNP is not consistently altered in normal subjects in response to acute exercise [41, 83, 93–95]. However, with long-duration exercise, such as a 100 km ultramarathon, BNP levels were increased from resting values of 3.3–18.8 fmol/mL at the end of the race. The response of BNP to exercise is altered by a number of conditions [96]. When subjects performed submaximal exercise on a low-sodium diet, an increase in BNP was not noted; however, on a high-sodium diet, a significant increase was seen. A similar finding was reported with the presence or absence of fluid 217 intake in the course of exercise [83]. If subjects did not ingest water, there was no response to exercise, but if fluid was provided, BNP was increased with exercise. In hypertensive subjects, the increase in BNP with exercise was the same with or without beta-blockade, in contrast to the greater increase in ANP with beta-blockade [88]. This suggests that while similar mechanisms, such as atrial stretch, fluid intake, and sodium status, modify the response of both BNP and ANP to exercise, the neurological component present in the regulation of ANP is not an important factor for BNP. Urodilatin Urodilatin, a natriuretic hormone derived in the kidneys, has been suggested to play a role in the renal handling of sodium [97, 98]. Schmidt et al. assessed the response of urodilatin and ANP during bicycle ergometer exercise at 60% of maximum for 1 h [99]. Plasma ANP concentrations increased, and the excretion of urodilatin decreased; i.e., the hormones had a negative correlation. The decrease in urodilatin was associated with a reduction in the percent of the filtered sodium load excreted. As urodilatin increased, the amount of sodium lost also increased. These findings suggest a possible role in the regulation of sodium homeostasis during exercise that needs to be investigated further. Adrenomedullin Adrenomedullin is reported to have natriuretic and diuretic effects. Adrenomedullin is produced in the vascular endothelium and in smooth muscle cells. In humans, plasma concentrations are responsive to changes in blood volume [100, 101]. Furthermore, changes in adrenomedullin are correlated with changes in ANP and BNP in patients. In normotensive subjects, adrenomedullin concentrations in response to submaximal exercise of short duration were not altered, even though ANP and BNP levels were increased. In contrast, during maximal exercise, Tanaka and colleagues found adrenomedullin to be increased by 45% compared to at rest and to be negatively associated with systolic blood pressure [102]. Piquard et al. also reported that with acute maximal 218 exercise, adrenomedullin increased from resting levels of 15–29 pmol/L at the end of ­exercise [103]. Yet others have found adrenomedullin to be increased with submaximal exercise and decreased with maximal exercise [104, 105]. Therefore, further investigation is warranted to elucidate the responses and actions for adrenomedullin during exercise. Fluid and Electrolyte Regulation The management of fluids and electrolytes is a careful balance between loss of salts and water through sweat, shifts between body compartments, and conservation by the kidneys and replenishment through ingestion [106]. While some losses are tolerated during exercise, once critical levels are exceeded, there are decrements in performance. In order to avoid these reductions, a series of compensatory mechanisms are activated that have to work in concert to maintain the milieu, to optimize performance, and to avoid subsequent morbidities and mortality. Total Body Water During exercise there is a loss of TBW, predominately via sweating and in part from increased respiratory loss. The reduction of TBW is tolerated until a critical level is attained. The loss of TBW during exercise is equivalent to the reduction in total body mass over the period of exercise performance. Though this assumption has been questioned, there is still a strong relationship between the decrease in TBW and body mass [107–109]. During long-duration exercise, the reduction in TBW may exceed 5% of body mass. In a 70 kg person, this would equate to fluid loss of 3000–4000 mL [6, 8, 110]. In laboratory experiments, a reduction of more than 2% body mass has been shown to decrease performance [110]. In contrast during competitive endurance events, a reduction of greater than 4% body mass was demonstrated to have a decrement in performance [9]. Of note, even with free access to C. E. Wade water, a loss in TBW during exercise is observed. This water loss, in the presence of fluids to ingest, is referred to as voluntary dehydration [18, 111]. Voluntary dehydration represents about 20–30% of the total loss of body water during an activity, as 70–80% is replaced by supplemental intake over the period of exercise. During a marathon the average body mass loss was 2.3% even though fluids were available. Interestingly, in subjects finishing under 3 h, the loss was 3.1%, from 3 to 4 h 2.5%, and over 4 h 1.8% [112]. The ability to tolerate a greater decrease in TBW was inversely associated with finish time. These observations suggest that individuals who are successful in these events are able to tolerate a greater TBW loss and still perform at a high level. The loss of fluids sustained in the course of exercise is usually replaced in the subsequent 24 h [21, 29, 60, 113]. Irrespective of the TBW loss tolerance, at some point the loss of TBW will impact the performance of an individual. The loss of TBW during exercise is not equally distributed throughout the body or between body fluid compartments. Over the course of exercise, there is a redistribution of fluids among the various compartments of the body, with a pronounced reduction in plasma volume [59, 61, 65, 114]. The reduction in plasma volume during maximal acute exercise is 8–12%, resulting in a 5–7% decrease in blood volume. This shift of fluids from the vascular space to the extravascular space has been attributed in part to increases in endothelial permeability, which could possibly be modified within specific tissues by angiotensin II, vasopressin, and norepinephrine [115–117]. The decrease in blood volume is compensated for by an increase in cardiac output and a redistribution of blood flow [48, 49, 118, 119]. During the performance of exercise, the redistribution of fluids within the vascular compartment is required to meet the metabolic demands of active tissues and to dissipate the thermal load resulting from the increase in metabolism. This redistribution of flow is the result of increases in local vascular resistance, which is in part due to hormonal regulation, predominately by catecholamines, angiotensin II, and vasopressin. 13 Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise Sweating The principal means of fluid and electrolyte loss during exercise is in sweat. Sweating is essential to dissipate the increased thermal load incurred by the elevation of metabolism with exercise [120]. The density of sweat pores is highly variable among subjects, as is the magnitude of sweat produced due to the subjects’ level of training and prior adaptation and acclimation to a hot environment [90, 120–123]. The rate of fluid loss by sweating can be as high as 1500 mL/h [6, 18, 108, 124]. The magnitude of fluid loss in sweat is hormonally mediated by vasopressin [125, 126]. Circulating levels of vasopressin are positively associated with the rate and composition of sweat during exercise. The rate of sweating during exercise is coupled with the changes in plasma osmolality and volume, the primary mediators of vasopressin; thus, it has been difficult to separate cause and effect [118, 119, 127, 128]. However, local subcutaneous injection of vasopressin alters the rate and composition of sweat from glands exposed to an increase in local skin temperature [129]. Plasma vasopressin concentrations have been associated with sweat sodium concentration and osmolality, suggesting vasopressin promotes water conservation in the sweat gland [125, 130]. In addition, studies involving a possible role of catecholamines on sweat rate have resulted in conflicting findings [131, 132]. However, in a study of the effect of fluid intake, it was shown that the ingestion of a large volume, >3 L, was associated with an increase in sweating, reduction in plasma concentration of norepinephrine, and an increase in skin blood flow. In contrast, the opposite effects were seen with ingestion of a small volume, >0.5 L, during long-duration submaximal exercise in the heat. Therefore, an increase in catecholamines appears to be associated with a decrease in skin blood flow that results in a decrease in sweating. Sweat is composed of a significant amount of electrolytes [90, 120, 121, 133, 134]. Thus, during exercise the predominate means of the loss of electrolytes is through sweat. The concentration of sodium in sweat ranges from 20 to 135 mmol/L, 219 potassium from 3 to 35 mmol/L, and chloride from 10 to 100 mmol/L, in contrast to “normal” plasma concentrations (sodium 135–145 mmol/L, potassium 3.5–5.0 mmol/L, and chloride 96–106 mmol/L) [135]. While the levels of electrolytes in sweat are lower than in plasma, the losses are significant. At a sweat rate of 1.5 L/h at a sodium concentration of 60 mmol/L, a total of 90 mmol would be lost or 3% of total body sodium. As noted above, however, the concentrations of electrolytes in sweat are highly variable. Electrolyte concentrations of sweat are decreased as a result of training and heat acclimation [65, 90, 121]. The lower concentrations reduce the tonicity of the sweat and therefore facilitate evaporation and cooling. In a comparison of 10 min of acute maximal exercise to 60 min of submaximal exercise (60% of maximum workload), minimal differences in the electrolyte concentrations were noted: sodium 70 vs. 77 mmol/L, potassium 7.7 vs. 4.8 mmol/L, and osmolality 171 vs. 172 mOSM/L for maximal and submaximal exercise, respectively. The reductions in the sodium concentration of sweat appear to be in part mediated by aldosterone [121, 136]. Fluid and Electrolyte Intake Consumption is the primary means of replacing the fluid and electrolytes losses incurred during the course of exercise [18, 137, 138]. In the performance of long-duration exercise, 80% of the fluid lost in sweat is replaced by voluntary ingestion if free access to fluids is provided [108, 137]. The extent to which volume losses are replaced is dependent upon the composition of the ingested fluid [137–141]. In humans during extended exercise, the volume of fluid replacement appears to be closely regulated. In contrast, the replacement of electrolytes does not appear to be as closely titrated and is a by-product of normal nutrient intake. Takamata et al. suggested that 6–24 h after heavy exercise, salt appetite is increased in association with a decrease in plasma osmolality and sodium concentrations resulting from fluid intake [113]. Leshem et al. 220 monitored salt intake after exercise and found a voluntary increase of 50% in the amount of salt added to food [142]. Passe et al. assessed the acceptance of hypertonic saline fluids during exercise and reported an increase in palatability of a 60 mmol/lL sodium solution, suggesting a relationship between sensory reception, hedonic response, and drink composition in the replacement of electrolytes post exercise [143]. Replacement of electrolytes may be coupled with hunger and increase in salt appetite. In animal models salt appetite is strongly associated with angiotensin II; however, this proposed relationship has yet to be definitively demonstrated in humans [11, 144, 145]. As previously noted, the replacement of fluids is closely controlled over the course of exercise and thus readily adjusted for following exercise. This tight regulation is modulated by thirst, the subjective sensation to seek and drink fluids [144–147]. The subjective sensation of thirst can persist for hours after exercise [113]. As described earlier there is a level of voluntary dehydration that can be tolerated in the performance of long-­ duration exercise, but the majority, about 80%, of the fluid loss is replaced by drinking. The residual loss associated with the level of voluntary dehydration is usually replaced within 24 h [21, 29, 113]. This process is associated with a variety of factors, such as the increase in plasma osmolality and reduction in blood volume, both of which are closely tied to the regulation of numerous hormones. Immediately after exercise Takamata et al. found the subjective evaluation of thirst to be immediately reduced upon ingestion of fluids yet increased hours later in spite of plasma osmolality being reduced [113]. This increase in thirst was associated with an elevation of aldosterone and presumably angiotensin II [91, 147]. If the replacement fluid is water, plasma osmolality and sodium concentration can be decreased before blood volume loss is corrected, thus presenting conflicting regulatory mechanisms resulting in a reduction in thirst [148, 149]. Merry et al. reported the subjective sensation of thirst to be associated with an increase in osmolality during moderate exercise C. E. Wade under various levels of hydration in subject with different levels of fitness (Fig. 13.2) [27]. Osmolality was also related to an increase in vasopressin, suggesting a possible association between vasopressin and thirst. Keneflick et al. assessed the response of thirst during 1 h of walking at 50% of maximum on a treadmill in temperate (27 °C) or cold (4 °C) environments [150]. In the cold environment, the sensation of thirst was reduced by 40% and associated with lower levels of vasopressin, even though plasma osmolality was increased. The authors speculated that peripheral vasoconstriction increased central blood volume that was sensed as an actual increase in blood volume. This hypothesis is supported in part by the observation that immersion and dehydration, which increase and decrease central blood volume, respectively, alter thirst via volume-induced stimulation of the cardiopulmonary baroreceptors. Stimulation of these baroreceptors by an increase in volume results in decreased vasopressin and PRA and increased ANP [151]. In contrast dehydration causing a reduction in volume elicits the opposite responses [33]. The specific roles of these hormones in the regulation of thirst during and following exercise have yet to be clearly defined. The ingestion of fluid during the performance of exercise has been advocated to sustain performance [4, 6, 110]. To determine fluid replacement by water ingestion during exercise, Robinson et al. had subjects perform two bouts of exercise, one with and another without fluids, on a cycle ergometer for 1 h at 85% of their maximum oxygen uptake [133]. The subjects ingested 1.5 L of water to replace the fluid loss due to sweating, which resulted in a 60% decrease in the loss of body mass. The ingestion of fluid did not alter sweat rate, the increase in body temperature, or perceived exertion. Though plasma osmolality and sodium concentrations had a greater increase in the absence of water intake, no differences in vasopressin or angiotensin II were reported. These findings were confirmed by McConell et al. who stated that ingestion of fluids had little benefit on exercise of 1 h [152]. However, others have consistently 13 Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise shown hypohydration to impair performance. There is an absence of data as to whether someone exercising should drink “as much as tolerable,” “to replace the weight lost during exercise,” or “ad libitum”; thus, Noakes et al. had also questioned the effects of fluid hydration during exercise [153]. The role of hormones in this debate is even more difficult to evaluate. Rehydration is shown to attenuate the response of atrial natriuretic hormone, vasopressin, and PRA to exercise [24, 32, 66]. Furthermore, the role of these hormones in the modulation of thirst during exercise is confounded. At present the data supports maintenance of an adequate hydration status to avoid the adverse effects of dehydration. The means of achieving this, and the levels needed, have yet to be defined. In light of the present state of data in this area, an understanding of the function of hormones in the regulation of thirst is essential. Hew-Butler has reviewed the role of vasopressin in fluid balance and its possible role in dysnatremia, specifically exercise-associated hyponatremia [10]. Hyponatremia with exercise may result from water retention associated with excess fluid intake, sodium loss predominately via sweat, or more likely a combination of these factors. Put forth is the hypothesis that non-osmotic-­mediated AVP release from the pituitary increases circulating levels of vasopressin leading to retention of water, even if fluid intake does not exceed recommended guidelines. This inappropriate fluid retention/overload could be a contributing factor of hyponatremia and its subsequent sequelae. The efforts from this group, in the lab and in the field, provide insights as to the contribution of vasopressin and other hormones to the regulation of fluid and electrolyte homeostasis [10, 22, 26, 70, 71]. Renal Function The action of hormones in the regulation of kidney function is well defined due to their role in the pathophysiology of hypertension. While extensive studies have been directed at the study 221 of hormones on kidney function during exercise, the contribution of the kidneys to fluid and electrolyte balance is limited [59, 60, 154–157]. Zambraski described the limited contribution of the kidney noting that in a normal individual, the kidneys produce about 1 mL of urine a minute or 60 mL/h [53]. This is in comparison to the loss of fluid from sweat on the order of 1000–1500 mL/h, during moderate to heavy exercise. Zambraski estimated that during exercise the renal conservation of water would only account for 4% of the loss of water and about 8% for the sodium [53]. Thus, the conservation of fluid by the kidney is hampered by the limited amounts of water and electrolyte excreted in the basal state. Nevertheless, the hormonal influences on the kidney provide insights into their role in the overall maintenance of fluid and electrolyte homeostasis during and following exercise [53, 60, 158]. enal Blood Flow R At rest the kidney receives about 20% or approximately 1000 mL/min of the overall cardiac output. During exercise renal blood flow is reduced in relation to the intensity and duration of exercise. With mild to moderate exercise (50–70% of maximum workload), there are negligible changes, but with maximal exercise flow is decreased by 40–60% from the normal [45, 48, 53, 158–160]. The reduction in renal blood flow persists for over 1 h after completion of the exercise. This reduction is caused by vasoconstriction of afferent arterioles, associated with an increase in sympathetic nerve activity and circulating levels of norepinephrine derived from spillover from the kidney [45, 47, 53, 159, 161]. In animal models upon initiation of exercise, there is an immediate reduction in renal blood flow which increases over time to a steady state associated with the level of exercise [162]. This immediate decrease suggests the predominance of the neural regulatory component in the initial phase of exercise. The reduction in renal blood flow decreases the volume of fluid and electrolytes delivered to the glomeruli of the kidney and in turn contributes to regional shifts in renal blood flow within the kidneys. 222 lomerular Filtration Rate G The amount of fluid moving across the membrane of the glomeruli of the kidney is termed the glomerular filtration rate. The movement of fluid is the product of the drive pressure across the membrane and oncotic pressure of the plasma. As noted above there is an increase in afferent arteriole resistance with exercise; however, this is accompanied by an increase in efferent arteriole resistance facilitating filtration. The increase in efferent arteriole resistance is controlled by angiotensin II. Changes in the rate of glomerular filtration are related to the intensity and duration of exercise and may persist for up to 24 h after exercise [163, 164]. Minimal changes in filtration are observed with exercise of less than 50% of maximum. With acute maximal exercise or long-­ duration exercise above 70% of maximum, the rate of filtration may be decreased by 50–70%. With heavy exercise there is also an increase in the permeability of the glomerular membrane as demonstrated by the occurrence of an increase in protein excretion [53, 165]. This alteration of permeability is suggested to be in part mediated by norepinephrine, vasopressin, and angiotensin II and results in an increase in the excretion of protein [53, 163, 166, 167]. rine Flow Rate U Urine flow rate is the product of the amount of fluid filtered (glomerular filtration rate) and the net reabsorption of fluid in the tubules. With exercise of low intensity, there is either no change or a slight increase in urine flow rate [39, 155]. With acute maximal exercise or long-duration exercise eliciting voluntary dehydration, urine flow rates are decreased by 20–60% of the normal basal levels of 0.8–1.2 mL/min [53, 59, 60]. This minimal decrease results in the conservation of water in light of the losses due to sweating. The decrease in the amount of filtered water is predominately due to vasoconstriction of the afferent arterioles caused by norepinephrine [45, 48, 131, 161]. Exercise also causes an increase in the osmolality of urine, indicative of an increase in the reabsorption of water [57–59]. However, C. E. Wade decreases have been reported in urinary osmolality indicative of an increase in free water clearance during heavy exercise [53, 59]. Therefore the role of vasopressin in the control of water reabsorption in the collecting tubule during exercise has been questioned. There may be inhibition of vasopressin or the possibility of a “washout” of the osmotic gradient in the medullary area of the kidney due to the redistribution of blood flow associated with the actions of angiotensin II. After exercise the reduction in urine flow persists and may contribute to the rectification of fluid loss along with increased drinking [21, 113]. enal Handling of Electrolytes R At the normal rate of glomerular filtration, the amount of fluid equivalent to the TBW is filtered in 5–6 h. The filtrate contains electrolyte concentrations equivalent to those of plasma. Over the course of traversing through the kidneys, 80–99% of the filtered load of electrolytes is reabsorbed. This reabsorption is hormonally mediated for sodium and establishes an electrochemical gradient for the handling of other electrolytes and an osmotic gradient for the handling of other solutes. With acute exercise, the decrease in the excretion of electrolytes is predominately due to the reduction in glomerular filtration rate [21, 113, 156]. During and following long-duration exercise, the reabsorption of sodium is regulated by aldosterone [21, 113]. With daily heavy exercise, there is a persistent increase in aldosterone, which is strongly associated with an increase in the reabsorption of sodium (Fig. 13.3) [21]. In summary, with exercise, kidney function changes and is regulated by a number of hormonal systems. The major alterations effecting fluid and electrolyte homeostasis are a decrease in renal blood flow and an increase in the reabsorption of sodium. There are several fallacies as to the contribution of these changes in kidney function to the net maintenance of fluids and electrolytes. The primary misunderstanding is the quantitative contribution of the kidney to fluid balance and the roles of hormones in these changes. 13 Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise Summary Exercise elicits increases in a number of hormones important in the regulation of fluid and electrolyte homeostasis. The action of these hormones may persist for hours and days after completion of the exercise. While increases in hormone levels are noted, the regulation and actions of these hormones are often not well defined, specifically in relation to the changes in fluid and electrolyte balance during exercise. There are issues as to the influence by the type and duration of exercise on hormonal responses that are not often accounted for. Recent efforts employing multifactorial analysis are just beginning to define some of these factors. In addition, the role of hormones in the etiology of the detrimental effects of exercise, such as dehydration and dysnatremia, is beginning to be addressed. Finally, evidence is mounting to show that exercise plays a vital role in fluid and electrolyte homeostasis. Observations of the hormonal responses to exercise will lead to a better understanding of both exercise physiology and related disease processes. References 1. Wade CE, Freund BJ. Hormonal control of blood volume during and following exercise. In: Lamb DR, Gisolfi CV, editors. Perspectives in exercise science and sports medicine, vol. 3. Carmel: Benchmark; 1990. p. 207–41. 2. Viru A. Plasma hormones and physical exercise. Int J Sports Med. 1992;13(3):201–9. 3. Viru A. Hormones in muscular activity. Boca Raton: CRC; 1985. 4. Cheuvront SN, Carter R III, Sawka MN. Fluid balance and endurance exercise performance. Curr Sports Med Rep. 2003;2(4):202–8. 5. Coris EE, Ramirez AM, Van Durme DJ. Heat illness in athletes: the dangerous combination of heat, humidity and exercise. Sports Med. 2004;34(1):9–16. 6. Swaka MN, Franceconi RP, Young AJ. Influence of hydration level and body fluids on exercise performance in the heat. J Am Med Assoc. 1988;252:1165–9. 7. Sawka MN, Montain SJ, Latzka WA. Hydration effects on thermoregulation and performance in the heat. Comp Biochem Physiol A Mol Integr Physiol. 2001;128(4):679–90. 223 8. Murray B. Hydration and physical performance. J Am Coll Nutr. 2007;26(5 Suppl):542S–8S. 9. Goulet ED. Effect of exercise-induced dehydration on time-trial exercise performance: a meta-analysis. Br J Sports Med. 2011;45(14):1149–56. 10. Hew-Butler T. Arginine vasopressin, fluid balance and exercise: is exercise-associated hyponatraemia a disorder of arginine vasopressin secretion? Sports Med. 2010;40(6):459–79. 11. Stachenfeld NS. Acute effects of sodium ingestion on thirst and cardiovascular function. Curr Sports Med Rep. 2008;7(4 Suppl):S7–13. 12. Siegel AJ, d’Hemecourt P, Adner MM, Shirey T, Brown JL, Lewandrowski KB. Exertional dysnatremia in collapsed marathon runners: a critical role for point-of-care testing to guide appropriate therapy. Am J Clin Pathol. 2009;132(3):336–40. 13. Siegel AJ, Januzzi J, Sluss P, et al. Cardiac biomarkers, electrolytes, and other analytes in collapsed marathon runners: implications for the evaluation of runners following competition. Am J Clin Pathol. 2008;129(6):948–51. 14. Verbalis JG. Renal function and vasopressin during marathon running. Sports Med. 2007;37(4–5):455–8. 15. Fortney SM, Nadel ER, Wenger CB, Bove JR. Effect of blood volume on sweating rate and body fluids in exercising humans. J Appl Physiol. 1981;51(6):1594–600. 16. Brechue WF, Stager JM. Acetazolamide alters temperature regulation during submaximal exercise. J Appl Physiol. 1990;69(4):1402–7. 17. Zappe DH, Helyar RG, Green HJ. The interaction between short-term exercise training and a diuretic-­ induced hypovolemic stimulus. Eur J Appl Physiol Occup Physiol. 1996;72(4):335–40. 18. Greenleaf JE. The consequences of exercise on thirst and fluid intake. In: Ramsay DJ, Booth DA, editors. Thirst. London: Springer; 1991. p. 412–21. 19. Nagashima K, Wu J, Kavouras SA, Mack GW. Increased renal tubular sodium reabsorption during exercise-induced hypervolemia in humans. J Appl Physiol. 2001;91(3):1229–36. 20. Warburton DE, Gledhill N, Quinney HA. Blood volume, aerobic power, and endurance performance: potential ergogenic effect of volume loading. Clin J Sport Med. 2000;10(1):59–66. 21. Wade CE, Hill LC, Hunt MM, Dressendorfer RH. Plasma aldosterone and renal function in runners during a 20-day road race. Eur J Appl Physiol Occup Physiol. 1985;54(5):456–60. 22. Hew-Butler T, Jordaan E, Stuempfle KJ, et al. Osmotic and nonosmotic regulation of arginine vasopressin during prolonged endurance exercise. J Clin Endocrinol Metab. 2008;93(6):2072–8. 23. Freund BJ, Claybaugh JR, Hashiro GM, Buono M, Chrisney S. Exaggerated ANF response to exercise in middle-aged vs. young runners. J Appl Physiol. 1990;69(5):1607–14. 224 24. Khamnei S, Alipour MR, Ahmadiasl N. The combined effects of exercise and post dehydration water drinking on plasma arginine vasopressin, plasma osmolality and body temperature in healthy males. Int J Endocrinol Metab. 2005;2:80–6. 25. Mandroukas A, Metaxas TI, Heller J, et al. The effect of different exercise-testing protocols on atrial natriuretic peptide. Clin Physiol Funct Imaging. 2011;31(1):5–10. 26. Hew-Butler T, Noakes TD, Soldin SJ, Verbalis JG. Acute changes in arginine vasopressin, sweat, urine and serum sodium concentrations in exercising humans: does a coordinated homeostatic relationship exist? Br J Sports Med. 2010;44(10):710–5. 27. Merry TL, Ainslie PN, Walker R, Cotter JD. Fitness alters fluid regulatory but not behavioural responses to hypohydrated exercise. Physiol Behav. 2008;95(3):348–52. 28. Bentzen H, Pedersen RS, Nyvad O, Pedersen EB. Influence of training habits on exercise-induced changes in plasma atrial and brain natriuretic peptide and urinary excretion of aquaporin-2 in healthy man. Scand J Clin Lab Invest. 2002;62(7):541–51. 29. Wade CE, Dressendorfer RH, O’Brien JC, Claybaugh JR. Renal function, aldosterone, and vasopressin excretion following repeated long-distance running. J Appl Physiol. 1981;50(4):709–12. 30. Geelen G, Keil LC, Kravik SE, et al. Inhibition of plasma vasopressin after drinking in dehydrated humans. Am J Phys. 1984;247(6 Pt 2):R968–71. 31. Melin B, Jimenez C, Savourey G, et al. Effects of hydration state on hormonal and renal responses during moderate exercise in the heat. Eur J Appl Physiol Occup Physiol. 1997;76(4):320–7. 32. Khamnei S, Hosseinlou A, Ebrahimi H. The effect of volume of consumed water on drinking-induced sweating and plasma levels of arginine vasopressin, epinephrine and norepinephrine. Int J Endocrinol Metab. 2004;2(1):19–28. 33. Maresh CM, Gabaree-Boulant CL, Armstrong LE, et al. Effect of hydration status on thirst, drinking, and related hormonal responses during low-­ intensity exercise in the heat. J Appl Physiol. 2004;97(1):39–44. 34. De Souza MJ, Maresh CM, Maguire MS, Kraemer WJ, Flora-Ginter G, Goetz KL. Menstrual status and plasma vasopressin, renin activity, and aldosterone exercise responses. J Appl Physiol. 1989;67(2):736–43. 35. Stachenfeld NS, Taylor HS. Effects of estrogen and progesterone administration on extracellular fluid. J Appl Physiol. 2004;96(3):1011–8. 36. Stachenfeld NS, DiPietro L, Kokoszka CA, Silva C, Keefe DL, Nadel ER. Physiological variability of fluid-regulation hormones in young women. J Appl Physiol. 1999;86(3):1092–6. 37. Radke KJ, King KB, Blair ML, Fitzpatrick PG, Eldredge DH. Hormonal responses to the 6-minute walk test in women and men with coronary heart disease: a pilot study. Heart Lung. 2005;34(2):126–35. C. E. Wade 38. Stachenfeld NS, Gleim GW, Zabetakis PM, Nicholas JA. Fluid balance and renal response following dehydrating exercise in well-trained men and women. Eur J Appl Physiol Occup Physiol. 1996;72(5–6):468–77. 39. Freund BJ, Shizuru EM, Hashiro GM, Claybaugh JR. Hormonal, electrolyte, and renal responses to exercise are intensity dependent. J Appl Physiol. 1991;70(2):900–6. 40. Shim CY, Ha JW, Park S, et al. Exaggerated blood pressure response to exercise is associated with augmented rise of angiotensin II during exercise. J Am Coll Cardiol. 2008;52(4):287–92. 41. Kjaer A, Appel J, Hildebrandt P, Petersen CL. Basal and exercise-induced neuroendocrine activation in patients with heart failure and in normal subjects. Eur J Heart Fail. 2004;6(1):29–39. 42. Coiro V, Jotti GS, Volpi R, et al. Difference between diabetic and nondiabetic smokers in the pituitary response to physical exercise. Metabolism. 2004;53(9):1140–4. 43. Wolf JP, Nguyen NU, Dumoulin G, Berthelay S. Plasma renin and aldosterone changes during twenty minutes’ moderate exercise. Influence of posture. Eur J Appl Physiol Occup Physiol. 1986;54(6):602–7. 44. Perrault H, Melin B, Jimenez C, et al. Fluid-­ regulating and sympathoadrenal hormonal responses to peak exercise following cardiac transplantation. J Appl Physiol. 1994;76(1):230–5. 45. Tidgren B, Hjemdahl P, Theodorsson E, Nussberger J. Renal neurohormonal and vascular responses to dynamic exercise in humans. J Appl Physiol. 1991;70(5):2279–86. 46. Svedenhag J. The sympatho-adrenal system in physical conditioning. Significance for training-induced adaptations and dependency on the training state. Acta Physiol Scand Suppl. 1985;543:1–73. 47. Baer PG, McGiff JC. Hormonal systems and renal hemodynamics. Annu Rev Physiol. 1980;42:589–601. 48. Rowell LB. Human cardiovascular adjustments to exercise and thermal stress. Physiol Rev. 1974;54(1):75–159. 49. Boushel R. Muscle metaboreflex control of the circulation during exercise. Acta Physiol (Oxf). 2010;199(4):367–83. 50. Galbo H. Hormonal and metabolic adaptation to exercise. New York: Thieme-Stratton; 1983. 51. Galbo H, Holst JJ, Christensen NJ. Glucagon and plasma catecholamine responses to graded and prolonged exercise in man. J Appl Physiol. 1975;38(1):70–6. 52. Kotchen TA, Hartley LH, Rice TW, Mougey EH, Jones LG, Mason JW. Renin, norepinephrine, and epinephrine responses to graded exercise. J Appl Physiol. 1971;31(2):178–84. 53. Zambraski EJ. The kidney and body fluid balance during exercise. In: Buskirk ER, Puhl SM, editors. 13 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise Body fluid balance: exercise and sport. Boca Raton: CRC; 1996. p. 75–95. Peronnet F, Beliveau L, Boudreau G, Trudeau F, Brisson G, Nadeau R. Regional plasma catecholamine removal and release at rest and exercise in dogs. Am J Phys. 1988;254(4 Pt 2):R663–72. Meeusen R, Roelands B. Central fatigue and neurotransmitters, can thermoregulation be manipulated? Scand J Med Sci Sports. 2010;20(Suppl 3):19–28. Roelands B, Goekint M, Heyman E, et al. Acute norepinephrine reuptake inhibition decreases ­performance in normal and high ambient temperature. J Appl Physiol. 2008;105(1):206–12. Wade CE. Response, regulation, and actions of vasopressin during exercise: a review. Med Sci Sports Exerc. 1984;16(5):506–11. Weitzman R, Kleeman CR. Water metabolism and neurohypophyseal hormones. In: Maxwell MH, Kleeman CR, editors. Clinical disorders of fluid and electrolyte metabolism. New York: McGraw-Hill; 1980. p. 531–645. Wade CE, Claybaugh JR. Plasma renin activity, vasopressin concentration, and urinary excretory responses to exercise in men. J Appl Physiol. 1980;49(6):930–6. Wade CE, Freund BJ, Claybaugh JR. Fluid and electrolyte homeostasis during and following exercise: hormonal and non-hormonal factors. In: Claybaugh JR, Wade CE, editors. Hormonal regulation of fluids and electrolytes: environmental effects. New York: Plenum; 1989. p. 1–44. Convertino VA, Keil LC, Bernauer EM, Greenleaf JE. Plasma volume, osmolality, vasopressin, and renin activity during graded exercise in man. J Appl Physiol. 1981;50(1):123–8. Inder WJ, Hellemans J, Swanney MP, Prickett TC, Donald RA. Prolonged exercise increases peripheral plasma ACTH, CRH, and AVP in male athletes. J Appl Physiol. 1998;85(3):835–41. Saito T, Soya H. Delineation of responsive AVP-­ containing neurons to running stress in the hypothalamus. Am J Physiol Regul Integr Comp Physiol. 2004;286(3):R484–90. Burge J, Knechtle B, Knechtle P, Gnadinger M, Rust AC, Rosemann T. Maintained serum sodium in male ultra-marathoners—the role of fluid intake, vasopressin, and aldosterone in fluid and electrolyte regulation. Horm Metab Res. 2011;43(9):646–52. Convertino VA, Keil LC, Greenleaf JE. Plasma volume, renin, and vasopressin responses to graded exercise after training. J Appl Physiol. 1983;54(2):508–14. Montain SJ, Laird JE, Latzka WA, Sawka MN. Aldosterone and vasopressin responses in the heat: hydration level and exercise intensity effects. Med Sci Sports Exerc. 1997;29(5):661–8. Brandenberger G, Candas V, Follenius M, Libert JP, Kahn JM. Vascular fluid shifts and endocrine responses to exercise in the heat. Effect of 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 225 rehydration. Eur J Appl Physiol Occup Physiol. 1986;55(2):123–9. Melin B, Eclache JP, Geelen G, et al. Plasma AVP, neurophysin, renin activity, and aldosterone during submaximal exercise performed until exhaustion in trained and untrained men. Eur J Appl Physiol Occup Physiol. 1980;44(2):141–51. Coiro V, Maffei ML, Volta E, et al. Effect of serotonergic system on AVP secretion induced by physical exercise. Neuropeptides. 2010;44(1):53–6. Hew-Butler T, Dugas JP, Noakes TD, Verbalis JG. Changes in plasma arginine vasopressin concentrations in cyclists participating in a 109-km cycle race. Br J Sports Med. 2010;44(8):594–7. Hew-Butler T, Hoffman MD, Stuempfle KJ, Rogers IR, Morgenthaler NG, Verbalis JG. Changes in copeptin and bioactive vasopressin in runners with and without hyponatremia. Clin J Sport Med. 2011;21(3):211–7. Barron JL, Noakes TD, Levy W, Smith C, Millar RP. Hypothalamic dysfunction in overtrained athletes. J Clin Endocrinol Metab. 1985;60(4): 803–6. Bouissou P, Richalet JP, Galen FX, et al. Effect of beta-adrenoceptor blockade on renin-aldosterone and alpha-ANF during exercise at altitude. J Appl Physiol. 1989;67(1):141–6. Hespel P, Lijnen P, Vanhees L, Fagard R, Amery A. Beta-adrenoceptors and the regulation of blood pressure and plasma renin during exercise. J Appl Physiol. 1986;60(1):108–13. Reid IA, Ganong WF. Control of aldosterone secretion. In: Genest J, Koiw E, Kuchel O, editors. Hypertension pathophysiology and treatment. New York: McGraw-Hill; 1977. p. 265–92. Taverner D, Mackay IG, Craig K, Watson ML. The effects of selective beta-adrenoceptor antagonists and partial agonist activity on renal function during exercise in normal subjects and those with moderate renal impairment. Br J Clin Pharmacol. 1991;32(3):387–91. Tanaka H, Shindo M, Gutkowska J, et al. Effect of acute exercise on plasma immunoreactive-atrial natriuretic factor. Life Sci. 1986;39(18):1685–93. Gleim GW, Zabetakis PM, DePasquale EE, Michelis MF, Nicholas JA. Plasma osmolality, volume, and renin activity at the “anaerobic threshold”. J Appl Physiol. 1984;56(1):57–63. Morris DJ. The metabolism and mechanism of action of aldosterone. Endocr Rev. 1981;2(2):234–47. Wade CE, Ramee SR, Hunt MM, White CJ. Hormonal and renal responses to converting enzyme inhibition during maximal exercise. J Appl Physiol. 1987;63(5):1796–800. Niessner A, Ziegler S, Slany J, Billensteiner E, Woloszczuk W, Geyer G. Increases in plasma levels of atrial and brain natriuretic peptides after running a marathon: are their effects partly counterbalanced by adrenocortical steroids? Eur J Endocrinol. 2003;149(6):555–9. C. E. Wade 226 82. Freund BJ, Wade CE, Claybaugh JR. Effects of exercise on atrial natriuretic factor. Release mechanisms and implications for fluid homeostasis. Sports Med. 1988;6(6):364–77. 83. Kaka S, Mudambo MD, Coutie W, Rennie MJ. Plasma arginine vasopressin, atrial natriuretic peptide and brain natriuretic peptide responses to long-term field training in the heat: effects of fluid ingestion and acclimatization. Eur J Appl Physiol. 1997;75:219–25. 84. Freund BJ, Claybaugh JR, Dice MS, Hashiro GM. Hormonal and vascular fluid responses to ­maximal exercise in trained and untrained males. J Appl Physiol. 1987;63(2):669–75. 85. Ohba H, Takada H, Musha H, et al. Effects of prolonged strenuous exercise on plasma levels of atrial natriuretic peptide and brain natriuretic peptide in healthy men. Am Heart J. 2001;141(5):751–8. 86. Geny B, Charloux A, Lampert E, Lonsdorfer J, Haberey P, Piquard F. Enhanced brain natriuretic peptide response to peak exercise in heart transplant recipients. J Appl Physiol. 1998;85(6):2270–6. 87. Geny B, Richard R, Mettauer B, Lonsdorfer J, Piquard F. Cardiac natriuretic peptides during exercise and training after heart transplantation. Cardiovasc Res. 2001;51(3):521–8. 88. Tanaka M, Ishizaka Y, Ishiyama Y, et al. Chronic effect of beta-adrenoceptor blockade on plasma levels of brain natriuretic peptide during exercise in essential hypertension. Hypertens Res. 1996;19(4):239–45. 89. Wambach G, Koch J. BNP plasma levels during acute volume expansion and chronic sodium loading in normal men. Clin Exp Hypertens. 1995;17(4):619–29. 90. Cuneo RC, Espiner EA, Nicholls MG, Yandle TG, Joyce SL, Gilchrist NL. Renal, hemodynamic, and hormonal responses to atrial natriuretic peptide infusions in normal man, and effect of sodium intake. J Clin Endocrinol Metab. 1986;63(4):946–53. 91. Fitzsimons JT, Simons BJ. The effect on drinking in the rat of intravenous infusion of angiotensin, given alone or in combination with other stimuli of thirst. J Physiol. 1969;203(1):45–57. 92. Sudoh T, Minamino N, Kangawa K, Matsuo H. Brain natriuretic peptide-32: N-terminal six amino acid extended form of brain natriuretic peptide identified in porcine brain. Biochem Biophys Res Commun. 1988;155(2):726–32. 93. Yamada T, Nakao K, Morii N, et al. Central effect of atrial natriuretic polypeptide on angiotensin II-stimulated vasopressin secretion in conscious rats. Eur J Pharmacol. 1986;125(3):453–6. 94. Tanaka M, Ishizaka Y, Ishiyama Y, et al. Exercise-­ induced secretion of brain natriuretic peptide in essential hypertension and normal subjects. Hypertens Res. 1995;18(2):159–66. 95. Bentzen H, Pedersen RS, Nyvad O, Pedersen EB. Effect of exercise on natriuretic peptides in 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. plasma and urine in chronic heart failure. Int J Cardiol. 2004;93(2–3):121–30. Kato M, Kinugawa T, Ogino K, et al. Augmented response in plasma brain natriuretic peptide to dynamic exercise in patients with left ventricular dysfunction and congestive heart failure. J Intern Med. 2000;248(4):309–15. Gerzer R, Drummer C. Is the renal natriuretic peptide urodilatin involved in the regulation of natriuresis? J Cardiovasc Pharmacol. 1993;22(Suppl 2):S86–7. Kentsch M, Otter W, Drummer C, et al. The dihydropyridine calcium channel blocker BAY t 7207 attenuates the exercise induced increase in plasma ANF and cyclic GMP in patients with mildly impaired left ventricular function. Eur J Clin Pharmacol. 1995;49(3):177–82. Schmidt W, Bub A, Meyer M, et al. Is urodilatin the missing link in exercise-dependent renal sodium retention? J Appl Physiol. 1998;84(1):123–8. Nishikimi T, Saito Y, Kitamura K, et al. Increased plasma levels of adrenomedullin in patients with heart failure. J Am Coll Cardiol. 1995;26(6):1424–31. Ishimitsu T, Nishikimi T, Saito Y, et al. Plasma levels of adrenomedullin, a newly identified hypotensive peptide, in patients with hypertension and renal failure. J Clin Invest. 1994;94(5):2158–61. Tanaka M, Kitamura K, Ishizaka Y, et al. Plasma adrenomedullin in various diseases and exercise-­ induced change in adrenomedullin in healthy subjects. Intern Med. 1995;34(8):728–33. Piquard F, Charloux A, Mettauer B, et al. Exercise-­ induced increase in circulating adrenomedullin is related to mean blood pressure in heart transplant recipients. J Clin Endocrinol Metab. 2000;85(8):2828–31. Krzeminski K, Mikulski T, Kruk B, Nazar K. Plasma adrenomedullin response to maximal exercise in healthy subjects. J Physiol Pharmacol. 2003;54(2):225–32. Krzeminski K, Mikulski T, Nazar K. Effect of prolonged dynamic exercise on plasma adrenomedullin concentration in healthy young men. J Physiol Pharmacol. 2006;57(4):571–81. Latzka WA, Montain SJ. Water and electrolyte requirements for exercise. Clin Sports Med. 1999;18(3):513–24. Pugh LG, Corbett JL, Johnson RH. Rectal temperatures, weight losses, and sweat rates in marathon running. J Appl Physiol. 1967;23(3):347–52. Rogers G, Goodman C, Rosen C. Water budget during ultra-endurance exercise. Med Sci Sports Exerc. 1997;29(11):1477–81. Tam N, Nolte HW, Noakes TD. Changes in total body water content during running races of 21.1 km and 56 km in athletes drinking ad libitum. Clin J Sport Med. 2011;21(3):218–25. Sawaka MN, Montain SJ, Latzka WA. Body fluid balance during exercise-heat exposure. In: Buskirk ER, Puhl SM, editors. Body fluid balance: exercise and sport. Boca Raton: CRC; 1996. p. 139–57. 13 Hormonal Regulation of Fluid and Electrolyte Homeostasis During Exercise 111. Greenleaf JE, Sargent F II. Voluntary dehydration in man. J Appl Physiol. 1965;20(4):719–24. 112. Zouhal H, Groussard C, Minter G, et al. Inverse relationship between percentage body weight change and finishing time in 643 forty-two-kilometre marathon runners. Br J Sports Med. 2011;45(14):1101–5. 113. Takamata A, Mack GW, Gillen CM, Nadel ER. Sodium appetite, thirst, and body fluid regulation in humans during rehydration without sodium replacement. Am J Phys. 1994;266(5 Pt 2):R1493–502. 114. Harrison MH. Effects on thermal stress and exercise on blood volume in humans. Physiol Rev. 1985;65(1):149–209. 115. Padilla J, Simmons GH, Bender SB, Arce-Esquivel AA, Whyte JJ, Laughlin MH. Vascular effects of exercise: endothelial adaptations beyond active muscle beds. Physiology (Bethesda). 2011;26(3):132–45. 116. Rush JWE, Aultman CD. Vascular biology of angiotensin and the impact o physical activity. Appl Physiol Nutr Metab. 2008;33:162–72. 117. Whyte JJ, Laughlin MH. Review: the effects of acute and chronic exercise on the vasculature. Acta Physiol. 2010;199:441–50. 118. Fortney SM, Nadel ER, Wenger CB, Bove JR. Effect of acute alterations of blood volume on circulatory performance in humans. J Appl Physiol. 1981;50(2):292–8. 119. Fortney SM, Wenger CB, Bove JR, Nadel ER. Effect of hyperosmolality on control of blood flow and sweating. J Appl Physiol. 1984;57(6):1688–95. 120. Sato K. The physiology, pharmacology, and biochemistry of the eccrine sweat gland. Rev Physiol Biochem Pharmacol. 1977;79:51–131. 121. Kirby CR, Convertino VA. Plasma aldosterone and sweat sodium concentrations after exercise and heat acclimation. J Appl Physiol. 1986;61(3):967–70. 122. Jorgenson RJ, Salinas CF, Dowben JS, St John DL. A population study on the density of palmar sweat pores. Birth Defects Orig Artic Ser. 1988;24(2):51–63. 123. Brown MB, McCarty NA, Millard-Stafford M. High-sweat Na+ in cystic fibrosis and healthy individuals does not diminish thirst during exercise in the heat. Am J Physiol Regul Integr Comp Physiol. 2011;301(4):R1177–85. 124. Wingo JE, Low DA, Keller DM, Brothers RM, Shibasaki M, Crandall CG. Skin blood flow and local temperature independently modify sweat rate during passive heat stress in humans. J Appl Physiol. 2010;109(5):1301–6. 125. Fasciolo JC, Totel GL, Johnson RE. Antidiuretic hormone and human eccrine sweating. J Appl Physiol. 1969;27(3):303–7. 126. Saini J, Geny B, Brandenberger G, et al. Training effects on the hydromineral endocrine responses of cardiac transplant patients. Eur J Appl Physiol Occup Physiol. 1995;70(3):226–33. 127. Coiro V, Casti A, Volta E, et al. Naloxone decreases the inhibitory effect of ethanol on the release of 128. 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 227 arginine-vasopressin induced by physical exercise in man. J Neural Transm. 2009;116(9):1065–9. Weidmann P, Hasler L, Gnadinger MP, et al. Blood levels and renal effects of atrial natriuretic peptide in normal man. J Clin Invest. 1986;77(3):734–42. Jougasaki M, Wei CM, Aarhus LL, Heublein DM, Sandberg SM, Burnett JC Jr. Renal localization and actions of adrenomedullin: a natriuretic peptide. Am J Phys. 1995;268(4 Pt 2):F657–63. Gibinski K, Kozlowski S, Chwalbinska-Moneta J, Giec L, Zmudzinski J, Markiewicz A. ADH and thermal sweating. Eur J Appl Physiol Occup Physiol. 1979;42(1):1–13. Mack GW, Shannon LM, Nadel ER. Influence of beta-adrenergic blockade on the control of sweating in humans. J Appl Physiol. 1986;61(5):1701–5. Allen JA, Jenkinson DJ, Roddie IC. The effect of -adrenoceptor blockade on human sweating. Br J Pharmacol. 1973;47(3):487–97. Robinson TA, Hawley JA, Palmer GS, et al. Water ingestion does not improve 1-h cycling performance in moderate ambient temperatures. Eur J Appl Physiol Occup Physiol. 1995;71(2–3):153–60. Verde T, Shephard RJ, Corey P, Moore R. Sweat composition in exercise and in heat. J Appl Physiol. 1982;53(6):1540–5. Robinson S, Robinson AH. Chemical composition of sweat. Physiol Rev. 1954;34(2):202–20. Collins KJ. The action of exogenous aldosterone on the secretion and composition of drug-induced sweat. Clin Sci. 1966;30(2):207–21. Coyle EF, Hamilton M. Fluid replacement during exercise; effects on physiological homeostasis and performance. In: Lamb DR, Gisolfi CV, editors. Perspectives in exercise science and sports medicine, vol. 3. Carmel: Benchmark; 1990. p. 281–303. Nadel ER, Mack GW, Nose H. Influence of fluid replacement beverages on body fluid homeostasis during exercise and recovery. In: Lamb DR, Gisolfi CV, editors. Perspectives in exercise science and sports medicine, vol. 3. Carmel: Benchmark; 1990. p. 181–206. Evans GH, Shirreffs SM, Maughan RJ. Postexercise rehydration in man: the effects of carbohydrate content and osmolality of drinks ingested ad libitum. Appl Physiol Nutr Metab. 2009;34(4):785–93. Kavouras SA, Armstrong LE, Maresh CM, et al. Rehydration with glycerol: endocrine, cardiovascular, and thermoregulatory responses during exercise in the heat. J Appl Physiol. 2006;100(2):442–50. Fritzsche RG, Switzer TW, Hodgkinson BJ, Lee SH, Martin JC, Coyle EF. Water and carbohydrate ingestion during prolonged exercise increase maximal neuromuscular power. J Appl Physiol. 2000;88(2):730–7. Leshem M, Abutbul A, Eilon R. Exercise increases the preference for salt in humans. Appetite. 1999;32(2):251–60. Passe DH, Stofan JR, Rowe CL, Horswill CA, Murray R. Exercise condition affects hedonic C. E. Wade 228 144. 145. 146. 147. 148. 149. 150. 151. 152. 153. 154. 155. 156. responses to sodium in a sport drink. Appetite. 2009;52(3):561–7. Stricker EM, Verbalis JG. Hormones and behavior: the biology of thirst and sodium appetite. Am Scientist. 1988;76:261–7. Stricker EM, Huang W, Sved AF. Early osmoregulatory signals in the control of water intake and neurohypophyseal hormone secretion. Physiol Behav. 2002;76(3):415–21. Rolls BJ, Edmund TR. In: Gray J, editor. Thirst— problems in the behavioural sciences. Cambridge: Cambridge University Press; 1982. Szczepanska-Sadowska E. Hormonal inputs to thirst. In: Ramsay DJ, Booth DA, editors. Thirst. London: Springer; 1991. p. 110–30. Nose H, Mack GW, Shi XR, Nadel ER. Role of osmolality and plasma volume during rehydration in humans. J Appl Physiol. 1988;65(1):325–31. Nose H, Mack GW, Shi XR, Nadel ER. Involvement of sodium retention hormones during rehydration in humans. J Appl Physiol. 1988;65(1):332–6. Kenefick RW, Hazzard MP, Mahood NV, Castellani JW. Thirst sensations and AVP responses at rest and during exercise-cold exposure. Med Sci Sports Exerc. 2004;36(9):1528–34. Wada F, Sagawa S, Miki K, et al. Mechanism of thirst attenuation during head-out water immersion in men. Am J Phys. 1995;268(3 Pt 2):R583–9. McConell GK, Burge CM, Skinner SL, Hargreaves M. Influence of ingested fluid volume on physiological responses during prolonged exercise. Acta Physiol Scand. 1997;160(2):149–56. Noakes TD. Is drinking to thirst optimum? Ann Nutr Metab. 2010;57(Suppl 2):9–17. Castenfors J. Renal function during exercise. With special reference to exercise proteinuria and the release of renin. Acta Physiol Scand Suppl. 1967;293:1–44. Kachadorian WA, Johnson RE. Renal responses to various rates of exercise. J Appl Physiol. 1970;28(6):748–52. Poortmans JR. Exercise and renal function. Sports Med. 1984;1(2):125–53. 157. Poortmans JR, Vanderstraeten J. Kidney function during exercise in healthy and diseased humans. An update. Sports Med. 1994;18(6):419–37. 158. Zambraski EJ. Renal regulation of fluid homeostasis during exercise. In: Gisolfi CV, Lamb DR, editors. Perspectives in exercise science and sports medicine, vol. 3. Carmel: Benchmark; 1990. p. 247–76. 159. Suzuki M, Sudoh M, Matsubara S, Kawakami K, Shiota M, Ikawa S. Changes in renal blood flow measured by radionuclide angiography following exhausting exercise in humans. Eur J Appl Physiol Occup Physiol. 1996;74(1–2):1–7. 160. McAllister RM. Adaptations in control of blood flow with training: splanchnic and renal blood flows. Med Sci Sports Exerc. 1998;30(3):375–81. 161. Johnson MD, Barger AC. Circulating catecholamines in control of renal electrolyte and water excretion. Am J Phys. 1981;240(3):F192–9. 162. Zambraski EJ, Tucker MS, Lakas CS, Grassl SM, Scanes CG. Mechanism of renin release in exercising dog. Am J Phys. 1984;246(1 Pt 1):E71–6. 163. Poortmans JR, Mathieu N, De Plaen P. Influence of running different distances on renal glomerular and tubular impairment in humans. Eur J Appl Physiol Occup Physiol. 1996;72(5–6):522–7. 164. Tian Y, Tong TK, Lippi G, Huang C, Shi Q, Nie J. Renal function parameters during early and late recovery periods following an all-out 21-km run in trained adolescent runners. Clin Chem Lab Med. 2011;49(6):993–7. 165. Poortmans JR, Ouchinsky M. Glomerular filtration rate and albumin excretion after maximal exercise in aging sedentary and active men. J Gerontol A Biol Sci Med Sci. 2006;61(11):1181–5. 166. Lang CC, Rahman AR, Balfour DJ, Struthers AD. Effect of noradrenaline on renal sodium and water handling in euhydrated and overhydrated man. Clin Sci (Lond). 1993;85(4):487–94. 167. Bellinghieri G, Savica V, Santoro D. Renal alterations during exercise. J Ren Nutr. 2008;18(1): 158–64. Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone 14 Whitney R.D. Duff and Philip D. Chilibeck Introduction Adequate levels of physical activity are important for bone health. Proper exercise training increases bone mineral density and prevents osteoporosis and fractures [1]. Too much exercise without adequate energy replacement may however negatively affect hormone status (especially reproductive hormones), and this may have a negative impact on bone health. The intent of this chapter is to cover this spectrum of effects of exercise on bone physiology and health. The “Negative Effects of Exercise on Hormonal Regulation of Bone” section of this chapter covers the negative effects of high levels of exercise, especially without adequate dietary energy or calcium intake, on sex hormone (i.e., estrogen, testosterone) and calciotropic hormone (i.e., parathyroid hormone, calcitonin, and vitamin D) levels and how this can negatively impact bone health. The “Positive Effects of Exercise on Hormonal Regulation of Bone” section of this chapter covers how proper exercise training may be complimentary with sex hormones or may enhance anabolic and calciotropic hormones to improve bone health. Exercise may have additive or synergistic effects for increasing bone density when combined with estrogen replacement therapy in W. R.D. Duff · P. D. Chilibeck (*) University of Saskatchewan, College of Kinesiology, Saskatoon, SK, Canada e-mail: phil.chilibeck@usask.ca postmenopausal women. Although studies are mixed, exercising may induce acute increases in release of anabolic hormones (i.e., testosterone, growth hormone, insulin-like growth factor-1) or may alter release of calciotropic hormones (i.e., decrease parathyroid hormone, increase calcitonin and vitamin D). This may lead to changes in basal levels of these hormones with chronic training and improvement in bone health. egative Effects of Exercise N on Hormonal Regulation of Bone egative Effects of Exercise N on Reproductive Hormone Status Estrogen and Progesterone Estrogen and progesterone are important sex hormones for maintenance of bone health in women. Estrogen increases intestinal absorption of calcium [2] and decreases activity of bone-resorbing cells (osteoclasts) [3], whereas progesterone may positively affect cells involved in bone formation (osteoblasts) [4]. Excessive exercise without adequate energy replacement may have a negative impact on these hormones, resulting in amenorrhea, and negative impacts on bone health [5]. Athletic Amenorrhea Athletic amenorrhea (i.e., cessation of menses in premenopausal women) is most common in sports that require lower body mass [2–4], including long-distance © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_14 229 230 running [5] and rowing [6], and activities that involve subjective judgment [7], such as gymnastics [8], figure skating [9], and ballet [10]. Athletes in these sports may develop disordered eating in an attempt to maintain the lower body mass required. The “female athlete triad” was proposed as a diagnosis when disordered eating was present with amenorrhea and osteoporosis [7, 11]. Now considered a spectrum ranging from normal to varying degrees of pathology of the three components, the female athlete triad can be diagnosed when low energy availability (with or without disordered eating) is present along with a dysfunction in menstrual function and/or low bone mineral density [7, 12]. The modifications to the diagnoses guidelines increased overall prevalence of the disorder, although total prevalence is still unknown [4, 7]. Amenorrheic athletes have lower bone density than not only their eumenorrheic counterparts but also sedentary controls and have a two- to fourfold higher stress fracture rate [13, 14]. For some athletes however, high intensity of exercise, especially activities that involve high-impact forces or high strains on bone due to muscle pull, may offset some of the negative effects of amenorrhea. This may be the case in gymnasts [8, 15–17] and rowers [6]. Amenorrheic dancers may [18] or may not [10] have an elevated bone density at weight-bearing sites, but have an increased incidence of stress fractures [19]. Longer durations of menstrual dysfunction in dancers equate to larger deficits in bone density at the spine [14, 20, 21], with scoliosis forming in some [19]. Amenorrheic runners or endurance athletes generally have decreased bone density, even at weight-bearing sites [22–26], and this is associated with higher prevalence of stress fracture [27–31]. Weight-bearing exercise may therefore be of sufficient impact to be protective in gymnasts, but not in dancers and runners when normal menstruation is not maintained [26, 29]. Etiology of Athletic Amenorrhea Athletic amenorrhea has been attributed to increased cortisol levels due to chronic exercise stress or an imbalance between energy expenditure and W. R.D. Duff and P. D. Chilibeck energy intake (i.e., decreased energy availability), leading to reduced gonadotropin-releasing hormone pulse generation at the hypothalamus, subsequent decreased release of follicle-­ stimulating and luteinizing hormone from the anterior pituitary during the follicular phase, and reduction in the production of estrogen and progesterone in the luteal phase [2, 7]. Considering those on the spectrum with less severe menstrual dysfunction (i.e., not complete amenorrhea), a blunted release of these hormones still occur due to luteal phase defects [32]. The evidence for both hypotheses has previously been outlined by Loucks et al. [33] (see Chap. 11 in this book). The “stress hormone” hypothesis is supported by findings that high growth hormone levels and hypercortisolemia are often reported in amenorrheic athletes indicating alterations in the hypothalamic-­pituitary-adrenal axis [21, 34–39] and centrally driven increases in corticotropin-­ releasing factors that negatively affect gonadotropin secretion in animals and humans [35, 36, 39–42]. The “low energy availability” hypothesis is strongly supported by the literature. Firstly, amenorrheic and eumenorrheic athletes have lower dietary energy intakes relative to energy expenditure [43, 44] and have endocrine profiles (i.e., reduced levels of thyroid hormones; Fig. 14.1) [43, 45, 46] that occur during chronic energy deficiency [47]. Further, increased levels of the starvation hormone adiponectin, signaling “low energy availability” [39, 48], have been found in gymnasts and ballet dancers [39, 49]. Leptin, with physiological levels proportional to fat mass, could be considered the mediator hormone between energy availability and reproduction [50]. Low fat mass results in decreased production of leptin and increased production of ghrelin in amenorrheic athletes [2, 37, 39, 51– 55]; this suppresses the hypothalamic-pituitary-­ gonadal (HPG) axis leading to decreased estrogen levels [2, 39, 56]. Finally, short-term induction of menstrual cycle changes with exercise can be prevented by adequate dietary compensation [33] or administration of leptin [57]. “Low energy availability” (i.e., negative energy balance) and 14 Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone a 2 Triiodothyronine levels (nmol/L) b Thyroxine levels (nmol/L) 100 * 1.6 80 1.2 60 0.8 40 0.4 20 0 EU AM ANX 231 0 * EU AM ANX Fig. 14.1 EU eumenorrheic athletes; AM amenorrheic athletes; ANX anorexics. Amenorrheic athletes have endocrine profiles (i.e., decreased thyroid hormones) similar to anorexics with chronic energy deficiency. (Data taken from Refs. [43, 47]). ∗Eumenorrheic means are significantly different from amenorrheic and anorexic means (p < 0.05) the effect on the hypothalamus-pituitary-ovarian axis are described in detail by Stafford et al. [2] (summarized in Fig. 14.2). The decrease in bone mineral density with athletic amenorrhea is thought to be attributable to low energy availability affecting bone turnover, with resorption favored over formation [50]. It is therefore suggested that athletes may be able to reverse menstrual disorders and prevent bone loss without decreasing their energy expenditure (i.e., physical activity levels) by increasing their dietary caloric intake [33]. This is supported by studies where energy availability is decreased either by energy restriction or by increased exercise. Low energy availability induced by energy restriction or increased exercise reduced leptin and insulin-­ like growth factor-1 (IGF-1) levels, but only energy restriction decreased markers of bone formation [58]. studies showed estrogen therapy to induce an overall statistically significant increase of 3.3% at the lumbar spine site only [59]. Confounding factors such as spontaneous resumption of menses and weight gain may influence the changes in bone density versus estrogen per se. Estrogen likely has no effect on metabolic factors that impair bone formation, but rather controls bone resorption, which may not necessarily be elevated in amenorrheic athletes [13, 60, 61]. As such, amenorrheic dancers and runners who resumed irregular menses and/or gained weight had larger gains in spine bone density (6.3–17%) over 15–24 months compared to those who achieved neither [62–64]. Further, weight gain was shown to independently predict bone density gains in oligo- and amenorrheic runners, although dietary calcium intake did as well [65]. Considering that clinical findings of estrogen therapy do not strongly support use for improving bone health in amenorrheic athletes [13, 50, 59], clinicians tend to recommend calcium and vitamin D supplementation [66]; however, the latter also has insufficient evidence in amenorrheic athletes, and there is no consensus on appropriate dosage in this population [13]. Thus, the “cornerstone” of treatment has been identified as improving energy availability by increasing caloric intake while maintaining energy expenditure, resulting in weight gain and Prevention and Treatment of Athletic Amenorrhea Previous pharmacologic strategies to improve bone health in hypothalamic amenorrhea have included estrogen therapy and calcium supplementation. Estrogen therapy has shown mixed results, with some studies showing improvements or maintenance of bone density and with several others showing inconclusive results [13]. A recent systematic review and meta-analysis of controlled and noncontrolled W. R.D. Duff and P. D. Chilibeck 232 Fig. 14.2 The hypothalamic-pituitary-­ ovarian axis. Energy imbalance causes hypoestrogenism and amenorrhea. Decreases in leptin and increases in ghrelin may influence gonadotropin-releasing hormone (GnRH) secretion causing subsequent decreases in luteinizing hormone (LH) and estrogen production. (Adapted from Stafford et al. [2]) Negative energy balance Leptin Ghrelin Hypothalamus Abnormal GnRH pulse Anterior Pituitary LH secretion Ovaries Estrogen Bone ( resorption) hopefully resumption of regular menses [7, 12, 13, 33, 50, 67, 68]. Despite improvements with these treatments, bone density often still remains lower than control levels in many formerly amenorrheic athletes [2, 32, 62, 63, 69, 70]. This suggests adolescent athletes with amenorrheainduced low bone mineral content may experience difficulty with “catch-up” accrual [22, 71] and may develop premenopausal osteopenia and be at higher risk of osteoporotic fractures later in life [2, 13]. Even elite amenorrheic gymnasts showed compromised skeletal health after retirement [72, 73]. Prevention of irreversible bone loss due to reproductive, stress, and metabolic hormone dysregulation as a result of energy deficiency is prioritized over treatment [39, 70, 74]. Treatment is difficult as athletes will be resistant [46], but regardless should consider multiple individualized factors [4], and utilize a comprehensive assessment by a multidisciplinary team that includes a physician, dietitian, and psychologist [2, 75, 76]. Testosterone Excessive exercise without adequate energy replacement may also affect sex hormone status in men [77–80]. Adequate testosterone levels are important for proper calcium absorption [81] and stimulation of osteoblasts and therefore bone formation [82]. Reduced concentrations of free and total testosterone in response to chronic endurance exercise have been deemed the “exercise-­ hypogonadal male condition” [79]. More recently, a parallel to the female athlete triad has been suggested in males engaged in sports that emphasize leanness or weight control, with the triad including low energy availability (with or without disordered eating), hypogonadotropic 14 Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone hypogonadism, and low bone mineral density [83]. Disruption in the hypothalamic-pituitary-­ testicular axis with excessive exercise in males [79] may have similar etiology as dysregulations seen in females, owing to decreased energy availability [78] or production of stress hormones [84–87]. It is suggested however that the HPG axis may be less sensitive to physical stress and more sensitive to disordered eating in males [88]. A few studies, but not all, have successfully linked low energy availability to the suppression of the HPG axis in men [31, 89–91]. Previous evidence supports this link, such that a shift in caloric balance following a season in wrestlers allowed for an increase in body weight, returning testosterone levels to normal [78]. However, the available research on this topic in men is extremely limited compared to that which has studied women. Simple measures of hormone levels, such as testosterone, that influence reproduction have been relied upon to determine hypogonadotropic hypogonadism in males, since clinical determination would require complicated techniques such as sperm and fertility analyses, rather than simply a lack of menses to determine the female alternative of amenorrhea [83]. Cross-sectional studies have shown testosterone levels are lower in endurance- and resistance-trained men compared to controls [77, 78, 89, 90, 92–96], while studies also demonstrate testosterone suppression after periods of high volume training [97– 103]. Notably, some studies showed a lack of elevation of luteinizing hormone corresponding to suppressed testosterone [77, 93, 103] which may be attributed to deficiency of gonadotropin-­ releasing hormone, as seen in female athletes [104, 105]. Further, male athletes who participate in low- or no-impact and weight-class sports are at higher risk of impaired bone health, although a representative prevalence remains unknown [83]. These observations may imply a connection between training, suppressed testosterone, and impaired bone health. This is evident in a couple of studies where reduced testosterone in male cyclists [106] or runners [31], attributable to low energy availability, was associated with lower lumbar spine bone mineral density or increased 233 fractures. However, the association between testosterone levels and bone health is less clear in other studies. Some studies show that while male runners [88, 107–111] and cyclists [112–114] have reduced bone mass, primarily at the lumbar spine, testosterone levels are normal [88, 107, 110, 112]. To add to these observations in runners, in one study a negative association between training volume and bone density with no difference in testosterone levels was shown [110], while another study showed a negative association between training volume and testosterone levels with bone density unaffected [115]. One useful study that assessed testosterone, luteinizing hormone, and bone density demonstrated that after 5 months of training, triathletes (classified as endurance athletes) had higher bone density despite lower testosterone levels without elevated luteinizing hormone than controls [103]. Finally, a very recent study noted that resistance-trained runners had higher bone density at all sites than nonresistance-trained runners and controls, with no differences in testosterone or any bone biomarkers except vitamin D (which was higher); these authors concluded the benefits for bone were therefore attributable to chronic loading of the bone and not physiologically modulated by low testosterone [104]. Reduced levels of androgens in males have rarely been linked to corresponding reductions in bone mass, possibly because testosterone levels remain within the normal range, albeit usually at the extreme low end (unlike estrogen in females, which fall below the normal range) [61, 79, 88]. In one case study, a male with hypogonadism, reduced bone mass, and skeletal fragility had testosterone levels return to normal after treatment with clomiphene citrate which stimulates gonadotropin secretion [116]. In a 4-year exercise intervention in middle-aged men, Remes et al. [117] reported significant associations between estradiol and testosterone and bone turnover markers at baseline, although only associations between estradiol and bone formation were significant at the 1-year and post-intervention mark. More recently it was shown that estradiol levels in male athletes, rather than testosterone, predicted bone mineral density, although, notably, testosterone 234 predicted estradiol levels [88]. Further research is needed to confirm which factors of the triad affects the hypothalamic-pituitary-testicular axis the greatest. Such research should focus on alterations in estrogens rather than androgens in males [83] and include metabolic hormones leptin and ghrelin [79]. egative Effect of Exercise N on Calciotropic Hormones The calciotropic hormones (parathyroid hormone, calcitonin, and to an extent, 1,25-­dihydroxyvitamin D3 [vitamin D]) are involved in calcium homeostasis and bone metabolism [118]. Parathyroid hormone is released from the parathyroid gland in response to low blood calcium levels. Parathyroid hormone stimulates osteoclasts to resorb bone so that blood calcium levels can be restored [119]. Calcitonin has a less powerful but opposite effect. Calcitonin is released from the thyroid gland when blood calcium levels are high and inhibits bone resorption. Vitamin D stimulates active intestinal calcium absorption. High levels of exercise may alter secretion of these calciotropic hormones, negatively affecting bone mineral status. egative Effects of Exercise N on Parathyroid Hormone Although parathyroid hormone is mainly recognized for its role in bone resorption, it has also been known to have a role in bone formation [120]. Whether parathyroid hormone has catabolic or anabolic effects depends on the mode of administration (when given as a pharmaceutical), signaling mechanism, and duration of exposure [121, 122], with continuous infusion stimulating bone resorption and intermittent exposure stimulating bone formation [123, 124]. Exercise is another important mediator of parathyroid hormone that is dependent on exercise duration and intensity [123]. Thus, studies of the effect of exercise on parathyroid hormone and bone responses are mixed. The studies demonstrating negative or no effects are discussed below, with W. R.D. Duff and P. D. Chilibeck studies demonstrating positive effects discussed later in the chapter. A study in mice utilizing an acute exercise bout (30-minute running) led to a twofold increase in systemic parathyroid hormone [125]. The response of parathyroid hormone to acute exercise bouts in humans is quite variable with studies reporting an increase in release [121, 126–132], a decrease in release [133], and no change [134–136]. One study in particular compared acute bouts performed at 15% above or below ventilatory threshold; parathyroid hormone was only increased after the higher intensity bout, suggesting a stimulation threshold [137]. Thus, intensity as well as duration, type of exercise, and recovery may influence parathyroid response, accounting for the discrepancies between studies [123]. Acute bouts of higher-­ intensity exercise are thus likely to increase release, and in this case, parathyroid may promote an anabolic effect on bone by increasing osteoblast response to mechanical loading [118, 128, 132, 138]. In response to an exhaustive acute exercise session, there were no differences between trained endurance athletes and recreationally active athletes, with both groups showing a postexercise increase in parathyroid hormone, as well as markers of bone turnover; thus, training status does not appear to influence the response to acute exercise [121, 139]. Aside from exercise variables, parathyroid response to acute exercise bouts can also be affected by calcium concentrations, acidosis, catecholamines, and training [123]. Acute exercise bouts cause decreases in serum ionized calcium concentrations, possibly through dermal losses via sweating or increased urinary calcium [140]. This acute decrease in ionized calcium may stimulate release of parathyroid hormone and an increase in bone resorption [140]. This implies acute release of parathyroid hormone is deleterious, in contrast to the studies mentioned above. Consuming calcium before exercise sessions, or injection of calcium during exercise sessions, may attenuate this effect [140, 141]. Two short-term (6–8 weeks) training studies have shown no changes in parathyroid hormone, with no substantial changes in bone resorption 14 Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone 235 [142, 143]. However, the response of bone formation was different between these studies, with the study in young women showing increases [143] and the study in older men showing decreases [142]. These differences could be attributed to the population study (age, sex), as well as the training type. One longitudinal study also showed no changes in parathyroid hormone or bone resorption after a 7-month triathlon season, with increases in bone formation and subsequent gains in lumbar spine bone density [144]. On the other hand, excessive chronic high-­ intensity exercise training may cause an increase in the continuous release of parathyroid hormone [126] and a deleterious effect on bone. This may be related to increases in stress hormones, such as catecholamines. Parathyroid hormone release is stimulated by catecholamines in animal models [145]; this correlates with the intensity [146] or volume [126] of exercise. Two longitudinal exercise training studies indicated elevated basal levels of parathyroid hormone, which were associated with increased bone turnover and reduced bone mineral [147, 148]. This effect is not consistent however, as further training resulted in a decrease in parathyroid hormone and an increase in bone mineral [148], and in another study, training resulted in elevated parathyroid hormone levels and an increase in bone mineral [149]. There may be an interaction between different hormone systems that affect the set point at which parathyroid hormone is released, with a decrease in estrogen (as seen with chronic overtraining) resulting in increased parathyroid hormone release [134]. Low estrogen levels in young overtrained athletes may amplify the effects of parathyroid hormone on bone turnover, similar to what is seen in postmenopausal women [150]. release [134]. This may avert the beneficial effect of calcitonin on preventing bone resorption in the subset of runners with low bone mass. Studies assessing vitamin D levels in athletes demonstrate that concentrations vary greatly [151] and can be influenced by the time of year and sunlight exposure, as well as diet and other lifestyle choices [152]. Vitamin D levels may be lower in runners with low bone mass (i.e., amenorrheic athletes) in comparison to eumenorrheic athletes and controls [27], although levels were still within a normal range. Male cyclists (i.e., non-weight bearing) have also been shown to have low bone density coincident with low vitamin D status [106, 112, 153]; this may be related to low energy availability [106]. The majority of recent studies show that athletes are deficient or insufficient in vitamin D, with deficiencies tending to exist in winter months; therefore, effect of season may have a greater influence on vitamin D status that is compounded by excessive exercise with inadequate dietary intake [154–162]. Similar to what occurs in hypoestrogenism, the stimulus of loading the bone may override the negative effect of vitamin D deficiency [151]. Indeed, a large cross-sectional study examined male athletes of differing sports, ages, and ethnicities and found no association between bone density and vitamin D deficiency [163], and a study in female synchronized swimmers showed that although vitamin D and insulin-like growth factor-1 (IGF-­1) levels were suppressed, bone ultrasound measurements and markers of bone turnover were not different compared to controls [164]. egative Effects of Exercise N on Calcitonin and Vitamin D Excessive levels of exercise may negatively impact calcitonin and vitamin D levels. Female runners with low bone mass had decreased calcitonin release in response to elevated blood calcium levels following exercise, whereas runners with normal bone mass had increased calcitonin I nteractions Between Exercise and Estrogen for Increasing Bone Mass ositive Effects of Exercise P on Hormonal Regulation of Bone Exercise and estrogen replacement may be complimentary therapies for increasing bone mass in postmenopausal women. When the two are ­combined, their effects on some bone sites may 236 be synergistic (i.e., greater than the addition of each therapy alone). Animal studies have demonstrated either additive [165] or synergistic [166, 167] effects with the two therapies in postmenopausal models. The majority of studies in postmenopausal women have shown exercise and estrogen replacement therapy to have an additive [168, 169] or synergistic [170] effect on bone mass of the spine and a synergistic effect on whole-body bone mass [168, 170]. One study found a synergistic effect on bone density at all sites measured (hip, spine, and total body) [171]. Another study found individual benefits on bone density at the spine, but no synergistic effects [172]. However, a recent meta-analysis showed hormone replacement therapy in combination with exercise training had greater benefits for bone density at the femoral neck and lumbar spine than exercise alone, seemingly confirming a synergistic effect [173]. Despite this, women have become hesitant to utilize hormone replacement therapy for bone health due to safety concerns [174]; thus, interest in phytoestrogens as alternative therapy has grown. Animal studies using postmenopausal models have shown cooperative effects of exercise and isoflavone (a plant-­based phytoestrogen) on bone density and properties at the hip, lumbar spine, and total body [175–177]. However, a recent study in postmenopausal women contradicted animal study findings, showing that either therapy alone maintained bone density of the total hip, but when the therapies were combined, there was a negative interaction that resulted in a decrease at the same site [178]. The differences between animal and human studies may reflect the signaling mechanism, with lower doses in humans activating primarily estrogen receptor-β which downregulates the detection of exercise loads, while higher doses in animals also activate estrogen receptor-α which increases proliferation of osteoblasts in response to loads [178]. Thus, this implies that estrogen may augment the response of bone to loading (or vice versa) with exercise and estrogen synergistically increasing bone mass when estrogen receptor-α is preferentially activated [179]. W. R.D. Duff and P. D. Chilibeck ffects of Exercise on Anabolic E Hormones Anabolic hormones, such as testosterone, growth hormone, and IGF-1 increase following acute exercise sessions, and basal levels of these hormones may also increase in response to chronic training. Synthesis of IGF-1 may be in conjunction with growth hormone, as its synthesis in the liver or other sites, such as muscle or bone, may be mediated by growth hormone [180]. Each of these anabolic hormones activates osteoblasts and therefore stimulates bone formation [82, 181, 182]. This section covers the effects of acute and chronic exercise on release of anabolic hormones and their potential for positively affecting the bone. cute Effects of Exercise on Anabolic A Hormones and Bone Metabolism Acute exercise sessions may stimulate increases in blood levels of anabolic hormones in both men and women. In men, a single bout of exercise has been shown to result in increases in growth hormone [183–186], IGF-1 [186, 187], and testosterone levels [80, 183, 184, 188, 189]. In women, growth hormone [190–192], IGF-1 [193], and testosterone [191, 194] levels also increased in response to acute exercise. Thus, similar responses to acute exercise occur in women, particularly regarding growth hormone [195–197], although the response seems to be attenuated with aging in both sexes [185, 198, 199]. The latter is likely due to insufficient exercise stimulus in older adults [200]. The exercise-induced response of growth hormone is well recognized and may be due to neural input, lactate, or nitric oxide, with pulsatile release amplified when “threshold” is attained [195]. It is presumed that hepatic secretion of IGF-1 is stimulated by the elevations in growth hormone [196]. However, increased production of IGF-1 has not always followed the same pattern as growth hormone changes [186, 187, 191, 192]; therefore, their release may be independent. The adrenal cortex releases testosterone, and this may be the mechanism by which testosterone levels in females are increased with exercise [194]. 14 Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone Several studies have related the increases in anabolic hormones with acute exercise to changes in markers of bone turnover. Repeated one-leg, knee-extension exercise resulted in an increase in serum growth hormone, with an exercise-induced uptake of growth hormone over the thigh and a release of IGF-1, in men and women with a simultaneous increase in markers of bone turnover [135]. A 30-minute cycling session in trained males increased serum growth hormone and IGF-1 [201], while biomarkers of bone turnover also increased [202]. Finally, high-force eccentric contractions in males induced increases in IGF-1 and makers of bone turnover [203]. This release of anabolic hormones and increase of bone turnover may result in increased bone formation with training, which may translate to enhanced bone mineral with long-term training. ffects of Exercise Training on Anabolic E Hormones and Bone Mass A high bone mass in some athletic groups may be associated with high basal levels of anabolic hormones. For example, young women involved with resistance training [204] or gymnastics training [205] have higher bone mass along with higher levels of IGF-1 compared to aerobically trained women and sedentary controls. In aerobically trained females, testosterone levels are significantly associated with bone density [206]. Endurance-trained postmenopausal women have higher bone density, IGF-1 levels, and a trend toward higher growth hormone levels than sedentary controls [207]. Alternatively, amenorrheic adolescent endurance athletes have lower bone mass and lower levels of IGF-1 levels than sedentary controls, with IGF-1 levels acting as an independent predictor of apparent lumbar bone density [25]. These cross-sectional studies suggest that exercise training may enhance basal anabolic hormone levels and stimulate bone formation. For premenopausal women, this may hold true as long as regular menses are maintained. Higher bone mass in athletes, however, is not always associated with increased anabolic hormone levels. Male masters athletes involved in speed-power events had greater bone mineral density than 237 endurance athletes and controls, but with no differences in testosterone or IGF-1 levels [208]. Also, male runners who participated in resistance training had higher bone mineral density than runners not participating in resistance training, but with no differences in testosterone [104]. Longitudinal training studies relating increases in anabolic hormones to increases in bone mineral density are mixed in their findings. Following a 7-month triathlon season, male triathletes had significant gains in lumbar spine bone density, with corresponding increases in IGF-1, although testosterone levels did not change [144]. Six months of aquatic exercise in postmenopausal women increased IGF-1 and growth hormone levels, along with enhancements in bone properties of the calcaneus, as assessed by ultrasound [209]. Twelve months of resistance or jump training in middle-aged men with low bone mass increased lumbar spine bone mineral density, bone formation markers (relative to resorption markers), and IGF-1 levels [210]. Other studies in humans assessing changes in anabolic hormones and bone health have demonstrated that beneficial effects of training on bone can be realized without changes in anabolic hormones. Eight weeks of resistance training in older men and women reduced markers of bone resorption without changes in IGF-1 levels [211]. Sixteen to 24 weeks of resistance training of middle-­aged or older men increased femoral neck or lumbar spine bone mineral density without changes in levels of testosterone, growth hormone, and IGF-1 [212, 213]. Likewise, gymnastics training produced significant increases in lumbar spine bone mineral density in young women [214] and calcaneus mechanical competence in pre- and peri-pubertal males [215] without a change in serum IGF-1 levels. ositive Effects of Exercise P on Calciotropic Hormones Parathyroid Hormone Parathyroid hormone stimulates bone resorption to maintain homeostasis when blood calcium levels are low [119] although several studies 238 have shown the parathyroid response is independent of calcium [127, 146, 216, 217]. With chronic exercise training, parathyroid hormone levels may be lowered [130, 207, 209]. This has been associated with higher bone mineral values: In cross-­sectional studies, male and female endurance-trained athletes have been found to have lower serum parathyroid hormone levels associated with higher bone mineral density when compared to inactive controls [130, 207]. Six months of aquatic exercise training in postmenopausal women reduced parathyroid hormone levels while enhancing bone structural properties at the calcaneus [209]. Rats endurance-­ trained by treadmill exercise also have lower parathyroid hormone levels and higher bone mass compared to untrained rats [218]. It is suggested that endurance training induces a new set point of parathyroid hormone release regulated by calcium, or permanently suppresses its release [118], since corresponding higher calcium concentrations were found with low parathyroid levels [130, 207]. More recently, mice were put through a short-term (21 days) training program, while parathyroid hormone was inhibited or increased. Parathyroid inhibition attenuated the structural-level mechanical property increases seen in placebotreated mice, while parathyroid enhancement increased trabecular and cortical bone volume with no effect on tissue- and structural-­ level mechanical properties as seen in placebo-treated mice [125] . In contrast to the above studies, an increase in basal levels of parathyroid hormone has been found following a resistance training program that increased bone mineral density in postmenopausal women [149]. As mentioned in the “Negative Effects of Exercise on Parathyroid Hormone” section, parathyroid hormone may have anabolic effects on bone through stimulation of osteoblasts, if released in an intermittent fashion [124]. Further research is needed to determine the exact direction of changes in basal parathyroid hormone levels in response to different training protocols and whether these changes can be considered beneficial or detrimental to bone. W. R.D. Duff and P. D. Chilibeck alcitonin and Vitamin D C Few studies have looked at the effects of exercise on calcitonin levels. In response to an acute exercise bout, calcitonin levels have been shown to increase [133]. Limited studies have determined the effects of exercise training, and those that did have shown inconsistent results. Short-term exercise training was shown to have no effect on serum calcitonin levels in one study [207], while other studies showed increased calcitonin levels [209, 219, 220]; this could prevent bone resorption. Cross-sectional studies indicate that vitamin D levels may be elevated in endurance-trained [207] and resistance-trained [104, 221] individuals, as well as decathletes [222, 223]. This is associated with a higher bone mass in some of these individuals compared to inactive controls [104, 207, 221, 222]. Rats trained by treadmill exercise have an increase in vitamin D levels, increased calcium balance, increased intestinal calcium absorption efficiency, and increased bone mass compared to untrained rats [218, 224]. Increases in growth hormone release with exercise training [135, 207] may simulate the production of the active form of vitamin D [225], resulting in increased intestinal calcium absorption [223] and increased bone mass [118]. Male triathletes showed increased vitamin D levels after a 7-month season, with gains in lumbar spine bone density [144]. While growth hormone was not measured in this study, IGF-1 increased post-season; this may also affect vitamin D production. Directions for Future Research Extreme exercise training negatively impacts bone owing to increased stress and changes in metabolic hormones (i.e., increased cortisol and ghrelin, reduced leptin), eventually suppressing the HPG axis and decreasing estrogen (in females) or testosterone (in males) production. This suppression manifests as athletic amenorrhea, in conjunction with the female athlete triad, in premenopausal women and has been researched a great deal. However, research on 14 Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone h­ypogonadotropic hypogonadism and the athletic triad in males is still lacking. Such future research in male athletes should focus on alterations in estrogens [83] and metabolic hormones [79] and include longitudinal follow-ups to determine if males also experience difficulty in “catchup” accrual of bone mineral. Further, longer-term studies determining the efficacy of treatment plans for the athletic triad that incorporate individualized factors and are delivered by a multidisciplinary team should be studied [2, 4, 75, 76]. Such treatment plans should focus on determining if improving energy availability can prevent reductions in reproductive hormones that may occur with chronic exercise. Research has consistently shown that acute exercise results in an increased anabolic hormone response in both men and women with corresponding changes in bone turnover. Further, cross-sectional data shows athletes have high basal anabolic hormone levels and bone mass. However, more research is required to understand the effects of exercise training on anabolic hormones and bone density. Such research should focus on the development of exercise prescriptions for optimal enhancement of long-term hormone profiles that result in bone formation. Evidence regarding vitamin D in relation to bone in athletes is quite consistent. Research on the other calciotropic hormones, calcitonin and parathyroid hormone, is lacking or inconsistent. Studies determining the effects of acute exercise and exercise training on calcitonin levels and bone are needed. Further research is needed to determine the response of parathyroid hormone to different training protocols and whether these changes can be considered beneficial or detrimental to bone. Summary Chronic exercise training without adequate energy replacement induces release of stress and metabolic hormones, which in turn suppress the hypothalamic-pituitary-gonadal axis and downregulate production of reproductive 239 hormones. These hormonal changes ultimately lead to low bone mineral density, which presents with low energy availability and a dysfunction in menstrual function (females) or hypogonadotropic hypogonadism (males) in the athletic triad [83]. In some cases, particularly in gymnasts, overloading the bone negates the deleterious effects of hypoestrogenism. Regardless, prevention strategies in males and premenopausal females should focus on early identification of those at risk of developing the athletic triad. Prevention is particularly pertinent because evidence suggests bone loss experienced in previously amenorrheic athletes is irreversible. If it is too late for prevention, the “cornerstone” of treatment is improving energy availability. Postmenopausal women can counteract deleterious effects of hypoestrogenism via hormone replacement therapy and exercise training, which has a synergistic effect for bone density at clinically relevant sites (i.e., femoral neck and lumbar spine). Athletes tend to be deficient in the calciotropic hormone vitamin D, particularly in winter months, with this effect compounded by low energy availability. However, the stimulus of bone loading may again override the negative effects of the deficiency. The effect of exercise on other calciotropic hormones (e.g., parathyroid hormone) is highly dependent on exercise variables. Due to a stimulation threshold, acute bouts of high-intensity exercise increase release of parathyroid hormone that results in an anabolic effect on bone, if calcium levels are adequate prior to exercise. Alternatively, chronic exercise training may decrease parathyroid hormone levels, which has been associated with higher bone mineral values. However, the set point at which parathyroid hormone is released is altered with changing estrogen levels and calcium concentrations. Anabolic hormones (i.e., testosterone, growth hormone, and IGF-1) increase in response to acute bouts of exercise in both men and women, although the response is attenuated with age likely due to insufficient exercise stimulus (see Chap. 23 in this book). Trained individuals have high basal levels of anabolic 240 hormones that may be associated with high bone mass, suggesting the changes in bone t­urnover with acute exercise may translate to improved bone health with long-term training. W. R.D. Duff and P. D. Chilibeck 13. Ducher G, Turner AI, Kukuljan S, Pantano KJ, Carlson JL, Williams NI, et al. Obstacles in the optimization of bone health outcomes in the female athlete triad. [Review]. Sports Med. 2011;41(7):587–607. 14. Pearce G, Bass S, Young N, et al. Does weight-­ bearing exercise protect against the effects of exercise-induced oligomenorrhea on bone density? References Osteoporos Int. 1996;6(6):448–52. 15. Fehling PC, Alekel L, Clasey J, Rector A, Stillman 1. Blimkie CJR, Chilibeck PD, Davison KS. Bone minRJ. A comparison of bone mineral densities among eralization patterns: reproductive endocrine, calcium female athletes in impact loading and active loading and physical activity influences during the lifespan. sports. Bone. 1995;17(3):205–10. In: Perspectives in exercise science and sports medi16. Bemben DA, Buchanan TD, Bemben MG, Knehans cine: exercise and the female—a life span approach. AW. Influence of type of mechanical loading, menCarmel: Cooper Publishing; 1996. p. 73–145. strual status, and training season on bone density 2. Stafford DEJ. Altered hypothalamic-pituitary-­ in young women athletes. J Strength Cond Res. ovarian axis function in young female ath2004;18(2):220–6. letes: implications and recommendations for 17. Helge EW, Kanstrup I-L. Bone density in female elite management. [Review] [47 refs]. Treat Endocrinol. gymnasts: impact of muscle strength and sex hor2005;4(3):147–54. mones. Med Sci Sports Exerc. 2002;34(1):174–80. 3. Sherman RT, Thompson RA. The female athlete 18. Young N, Formica C, Szmukler G, Seeman E. Bone triad. J Sch Nurs. 2004;20(4):197–202. density at weight-bearing and nonweight-bearing 4. Goodman LR, Warren MP. The female athlete and sites in ballet dancers: the effects of exercise, hypomenstrual function. [Review] [40 refs]. Curr Opin gonadism, and body weight. J Clin Endocrinol Obstet Gynecol. 2005;17(5):466–70. Metab. 1994;78(2):449–54. 5. Drinkwater BL, Nilson K, Chesnut CH, Bremner 19. Warren MP, Brooks-Gunn J, Hamilton LH, Warren WJ, Shainholtz S, Southworth MB. Bone mineral LF, Hamilton WG. Scoliosis and fractures in content of amenorrheic and eumenorrheic athletes. young ballet dancers. Relation to delayed menN Engl J Med. 1984;311(5):277–81. arche and secondary amenorrhea. N Engl J Med. 6. Wolman RL, Clark P, McNally E, Harries M, Reeve 1986;314(21):1348–53. J. Menstrual state and exercise as determinants of 20. Cann CE, Martin MC, Genant HK, et al. Decreased spinal trabecular bone density in female athletes. spinal mineral content in amenorrheic women. BMJ. 1990;301(6751):516–8. JAMA. 1984;251(5):626–9. 7. Matzkin E, Curry EJ, Whitlock K. Female athlete 21. Valentino R, Savastano S, Tommaselli AP, D’Amore triad: past, present, and future. [Review]. J Am Acad G, Dorato M, Lombardi G. The influence of intense Orthop Surg. 2015;23(7):424–32. ballet training on trabecular bone mass, hormone 8. Robinson TL, Snow-Harter C, Taaffe DR, Gillis D, status, and gonadotropin structure in young women. Shaw J, Marcus R. Gymnasts exhibit higher bone J Clin Endocrinol Metab. 2001;86(10):4674–8. mass than runners despite similar prevalence of 22. Barrack MT, Van Loan MD, Rauh MJ, Nichols amenorrhea and oligomenorrhea. J Bone Miner Res. JF. Body mass, training, menses, and bone in ado1995;10(1):26–35. lescent runners: a 3-yr follow-up. Med Sci Sports 9. Slemenda CW, Johnston CC. High intensity activiExerc. 2011;43(6):959–66. ties in young women: site specific bone mass 23. Rencken ML, Chesnut CH, Drinkwater BL. Bone effects among female figure skaters. Bone Miner. density at multiple skeletal sites in amenorrheic ath1993;20(2):125–32. letes. JAMA. 1996;276(3):238–40. 10. Warren MP, Brooks-Gunn J, Fox RP, Lancelot C, 24. Gremion G, Rizzoli R, Slosman D, Theintz G, Newman D, Hamilton WG. Lack of bone accretion Bonjour JP. Oligo-amenorrheic long-distance runand amenorrhea: evidence for a relative osteopenia ners may lose more bone in spine than in femur. Med in weight-bearing bones. J Clin Endocrinol Metab. Sci Sports Exerc. 2001;33(1):15–21. 1991;72(4):847–53. 25. Christo K, Prabhakaran R, Lamparello B, Cord J, 11. Otis CL, Drinkwater B, Johnson M, Loucks A, Miller KK, Goldstein MA, et al. Bone metabolism Wilmore J, American College of Sports Medicine in adolescent athletes with amenorrhea, athletes position stand. The female athlete triad. Med Sci with eumenorrhea, and control subjects. Pediatrics. Sports Exerc. 1997;29(5):i–ix. 2008;121(6):1127–36. 12. Nattiv A, Loucks AB, Manore MM, Sanborn CF, 26. Gibson JH, Harries M, Mitchell A, Godfrey R, Sundgot-Borgen J, Warren MP, et al. American Lunt M, Reeve J. Determinants of bone density and College of Sports Medicine position stand. The prevalence of osteopenia among female runners female athlete triad. Med Sci Sports Exerc. in their second to seventh decades of age. Bone. 2007;39(10):1867–82. 2000;26(6):591–8. 14 Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone 27. Marcus R, Cann C, Madvig P, Minkoff J, Goddard M, Bayer M, et al. Menstrual function and bone mass in elite women distance runners. Endocrine and metabolic features. Ann Intern Med. 1985;102(2):158–63. 28. Myburgh KH, Hutchins J, Fataar AB, Hough SF, Noakes TD. Low bone density is an etiologic factor for stress fractures in athletes. Ann Intern Med. 1990;113(10):754–9. 29. Joy EA, Campbell D. Stress fractures in the female athlete. [Review] [28 refs]. Curr Sports Med Rep. 2005;4(6):323–8. 30. Barrow GW, Saha S. Menstrual irregularity and stress fractures in collegiate female distance runners. Am J Sports Med. 1988;16:209–16. 31. Heikura IA, Uusitalo ALT, Stellingwerff T, Bergland D, Mero AA, Burke LM. Low energy availability is difficult to assess but outcomes have large impact on bone injury rates in elite distance athletes. Int J Sport Nutr Exerc Metab. 2018;28(4):403–11. 32. De Souza MJ. Menstrual disturbances in athletes: a focus on luteal phase defects. [Review] [45 refs]. Med Sci Sports Exerc. 2003;35(9):1553–63. 33. Loucks AB, Verdun M, Heath EM. Low energy availability, not stress of exercise, alters LH pulsatility in exercising women. J Appl Physiol 1985. 1998;84(1):37–46. 34. Ding JH, Sheckter CB, Drinkwater BL, Soules MR, Bremner WJ. High serum cortisol levels in exercise-associated amenorrhea. Ann Intern Med. 1988;108(4):530–4. 35. Loucks AB, Mortola JF, Girton L, Yen SS. Alterations in the hypothalamic-pituitary-­ ovarian and the hypothalamic-­ pituitary-adrenal axes in athletic women. J Clin Endocrinol Metab. 1989;68(2):402–11. 36. Laughlin GA, Yen SS. Nutritional and endocrine-­ metabolic aberrations in amenorrheic athletes. J Clin Endocrinol Metab. 1996;81(12):4301–9. 37. Laughlin GA, Yen SS. Hypoleptinemia in women athletes: absence of a diurnal rhythm with amenorrhea. J Clin Endocrinol Metab. 1997;82(1):318–21. 38. De Souza MJ, Maguire MS, Maresh CM, Kraemer WJ, Rubin KR, Loucks AB. Adrenal activation and the prolactin response to exercise in eumenorrheic and amenorrheic runners. J Appl Physiol 1985. 1991;70(6):2378–87. 39. Maimoun L, Georgopoulos NA, Sultan C. Endocrine disorders in adolescent and young female athletes: impact on growth, menstrual cycles, and bone mass acquisition. [Review]. J Clin Endocrinol. 2014;99(11):4037–50. 40. Mann DR, Jackson GG, Blank MS. Influence of adrenocorticotropin and adrenalectomy on gonadotropin secretion in immature rats. Neuroendocrinology. 1982;34(1):20–6. 41. Olster DH, Ferin M. Corticotropin-releasing hormone inhibits gonadotropin secretion in the ovariectomized rhesus monkey. J Clin Endocrinol Metab. 1987;65(2):262–7. 241 42. Williams CL, Nishihara M, Thalabard JC, Grosser PM, Hotchkiss J, Knobil E. Corticotropin-releasing factor and gonadotropin-releasing hormone pulse generator activity in the rhesus monkey. Electrophysiological studies. Neuroendocrinology. 1990;52(2):133–7. 43. Harber VJ, Petersen SR, Chilibeck PD. Thyroid hormone concentrations and muscle metabolism in amenorrheic and eumenorrheic athletes. Can J Appl Physiol. 1998;23(3):293–306. 44. Wilmore JH, Wambsgans KC, Brenner M, Broeder CE, Paijmans I, Volpe JA, et al. Is there energy conservation in amenorrheic compared with eumenorrheic distance runners? J Appl Physiol 1985. 1992;72(1):15–22. 45. Loucks AB, Laughlin GA, Mortola JF, Girton L, Nelson JC, Yen SS. Hypothalamic-pituitary-­ thyroidal function in eumenorrheic and amenorrheic athletes. J Clin Endocrinol Metab. 1992;75(2):514–8. 46. Birch K. Female athlete triad. [Review] [0 refs]. BMJ. 2005;330:244–6. 47. Harber VJ, Petersen SR, Chilibeck PD. Thyroid hormone concentrations and skeletal muscle metabolism during exercise in anorexic females. Can J Physiol Pharmacol. 1997;75(10–11):1197–202. 48. Ahima RS, Lazar MA. Adipokines and the peripheral and neural control of energy balance. Mol Endocrinol. 2008;22(5):1023–31. 49. Maïmoun L, Coste O, Philibert P, Briot K, Mura T, Galtier F, et al. Peripubertal female athletes in high-impact sports show improved bone mass acquisition and bone geometry. Metabolism. 2013;62(8):1088–98. 50. Warren MP, Chua AT. Exercise-induced amenorrhea and bone health in the adolescent athlete. [Review] [90 refs]. Ann N Y Acad Sci. 2008;1135:244–52. 51. Ackerman KE, Slusarz K, Guereca G, Pierce L, Slattery M, Mendes N, et al. Higher ghrelin and lower leptin secretion are associated with lower LH secretion in young amenorrheic athletes compared with eumenorrheic athletes and controls. Am J Physiol Endocrinol Metab. 2012;302(7):E800–6. 52. Christo K, Cord J, Mendes N, Miller KK, Goldstein MA, Klibanski A, et al. Acylated ghrelin and leptin in adolescent athletes with amenorrhea, eumenorrheic athletes and controls: a cross-sectional study. Clin Endocrinol. 2008;69(4):628–33. 53. Scheid JL, Toombs RJ, Ducher G, Gibbs JC, Williams NI, De Souza MJ. Estrogen and peptide YY are associated with bone mineral density in premenopausal exercising women. Bone. 2011;49(2):194–201. 54. Corr M, De Souza MJ, Toombs RJ, Williams NI. Circulating leptin concentrations do not distinguish menstrual status in exercising women. Hum Reprod. 2011;26(3):685–94. 55. De Souza MJ, Leidy HJ, O’Donnell E, al e. Fasting ghrelin levels in physically active women: relationship with menstrual disturbances and metabolic hormones. J Clin Endocrinol Metab. 2004;89:3536–42. 242 56. Ahima RS, Dushay J, Flier SN, Prabakaran D, Flier JS. Leptin accelerates the onset of puberty in normal female mice. J Clin Invest. 1997;99(3):391–5. 57. Welt CK, Chan JL, Bullen J, Murphy R, Smith P, DePaoli AM, et al. Recombinant human leptin in women with hypothalamic amenorrhea. N Engl J Med. 2004;351(10):987–97. 58. Papageorgiou M, Martin D, Colgan H, Cooper S, Greeves JP, Tang JCY, et al. Bone metabolic responses to low energy availability achieved by diet or exercise in active eumenorrheic women. Bone. 2018;114:181–8. 59. Altayar O, Al Nofal A, Carranza Leon BG, Prokop LJ, Wang Z, Murad MH. Treatments to prevent bone loss in functional hypothalamic amenorrhea: a systematic review and meta-analysis. J Endocr Soc. 2017;1(5):500–11. 60. Vescovi JD, Jamal SA, De Souza MJ. Strategies to reverse bone loss in women with functional hypothalamic amenorrhea: a systematic review of the literature. Osteoporos Int. 2008;19(4):465–78. 61. Bennell KL, Brukner PD, Malcolm SA. Effect of altered reproductive function and lowered testosterone levels on bone density in male endurance athletes. Br J Sports Med. 1996;30(3):205–8. 62. Drinkwater BL, Nilson K, Ott S, Chesnut CH. Bone mineral density after resumption of menses in amenorrheic athletes. JAMA. 1986;256(3):380–2. 63. Jonnavithula S, Warren MP, Fox RP, Lazaro MI. Bone density is compromised in amenorrheic women despite return of menses: a 2-year study. Obstet Gynecol. 1993;81(5 ( Pt 1)):669–74. 64. Warren MP, Brooks-Gunn J, Fox RP, Holderness CC, Hyle EP, Hamilton WG. Osteopenia in exercise-­ associated amenorrhea using ballet dancers as a model: a longitudinal study. J Clin Endocrinol Metab. 2002;87(7):3162–8. 65. Cobb KL, Bachrach LK, Sowers M, Nieves J, Greendale GA, Kent KK, et al. The effect of oral contraceptives on bone mass and stress fractures in female runners. Med Sci Sports Exerc. 2007;39(9):1464–73. 66. Carlson JL, Curtis M, Halpern-Felsher B. Clinician practices for the management of amenorrhea in the adolescent and young adult athlete. J Adolesc Health. 2007;40(4):362–5. 67. Mallinson RJ, Williams NI, Olmsted MP, Scheid JL, Riddle ES, De Souza MJ. A case report of recovery of menstrual function following a nutritional intervention in two exercising women with amenorrhea of varying duration. J Int Soc Sports Nutr. 2013;10:34. 68. De Souza MJ, Nattiv A, Joy E, Misra M, Williams NI, Mallinson RJ, et al. Female athlete triad coalition consensus statement on treatment and return to play of the female athlete triad: 1st international conference held in San Francisco, California, May 2012 and 2nd international conference held in Indianapolis, Indiana, May 2013. Br J Sports Med. 2014;48(4):289. W. R.D. Duff and P. D. Chilibeck 69. Drinkwater BL, Bruemner B, Chesnut CH. Menstrual history as a determinant of current bone density in young athletes. JAMA. 1990;263(4):545–8. 70. Keen AD, Drinkwater BL. Irreversible bone loss in former amenorrheic athletes. Osteoporos Int. 1997;7(4):311–5. 71. Wiksten-Almstromer M, Hirschberg AL, Hagenfeldt K. Reduced bone mineral density in adult women diagnosed with menstrual disorders during adolescence. Acta Obstet Gynecol Scand. 2009;88(5):543–9. 72. Ducher G, Eser P, Hill B, et al. History of amenorrhoe compromises some of the exercise-induced benefits in cortical and trabecular bone in the peripheral and axial skeleton: a study in retired elite gymnasts. Bone. 2009;29(45):760–7. 73. Eser P, Hill B, Ducher G, et al. Skeletal benefits after longterm retirement in former elite female gymnasts. J Bone Min Res. 2009;24(12):1981–8. 74. De Souza MJ, Williams NI. Beyond hypoestrogenism in amenorrheic athletes: energy deficiency as a contributing factor for bone loss. [Review] [59 refs]. Curr Sports Med Rep. 2005;4(1):38–44. 75. Hobart JA, Smucker DR. The female athlete triad. Am Fam Physician. 2000;61(11):3357–64, 3367. 76. Gabel KA. Special nutritional concerns for the female athlete. [Review] [40 refs]. Curr Sports Med Rep. 2006;5(4):187–91. 77. Wheeler GD, Wall SR, Belcastro AN, Cumming DC. Reduced serum testosterone and prolactin levels in male distance runners. JAMA. 1984;252(4):514–6. 78. Strauss RH, Lanese RR, Malarkey WB. Weight loss in amateur wrestlers and its effect on serum testosterone levels. JAMA. 1985;254(23):3337–8. 79. Hackney AC. Effects of endurance exercise on the reproductive system of men: the “exercise-­ hypogonadal male condition”. J Endocrinol Investig. 2008;31:932–8. 80. Eliakim A, Nemet D. Exercise and the male reproductive system. Harefuah. 2006;145(9):677–81, 702, 701. 81. Hope WG, Ibarra MJ, Thomas ML. Testosterone alters duodenal calcium transport and longitudinal bone growth rate in parallel in the male rat. Proc Soc Exp Biol Med. 1992;200(4):536–41. 82. Kasperk CH, Wergedal JE, Farley JR, Linkhart TA, Turner RT, Baylink DJ. Androgens directly stimulate proliferation of bone cells in vitro. Endocrinology. 1989;124(3):1576–8. 83. Tenforde AS, Barrack MT, Nattiv A, Fredericson M. Parallels with the female athlete triad in male athletes. Sports Med. 2016;46(2):171–82. 84. Cumming DC, Quigley ME, Yen SS. Acute suppression of circulating testosterone levels by cortisol in men. J Clin Endocrinol Metab. 1983;57(3):671–3. 85. Gambacciani M, Yen SS, Rasmussen DD. GnRH release from the mediobasal hypothalamus: in vitro inhibition by corticotropin-releasing factor. Neuroendocrinology. 1986;43(4):533–6. 14 Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone 86. Hackney AC. Endurance training and testosterone levels. Sports Med Auckl NZ. 1989;8(2):117–27. 87. Hackney AC. The male reproductive system and endurance exercise. Med Sci Sports Exerc. 1996;28(2):180–9. 88. Ackerman KE, Skrinar GS, Medvedova E, Misra M, Miller KK. Estradiol levels predict bone mineral density in male collegiate athletes: a pilot study. Clin Endocrinol. 2012;76(3):339–45. 89. Gomez-Merino D, Chennaoui M, Drogou C, Bonneau D, Guezennec CY. Decrease in serum leptin after prolonged physical activity in men. Med Sci Sports Exerc. 2002;34(10):1594–9. 90. De Souza MJ, Arce JC, Pescatello LS, Scherzer HS, Luciano AA. Gonadal hormones and semen quality in male runners. A volume threshold effect of endurance training. Int J Sports Med. 1994;15(7):383–91. 91. De Souza MJ, Miller BE. The effect of endurance training on reproductive function in male runners. A “volume threshold” hypothesis. Sports Med Auckl NZ. 1997;23(6):357–74. 92. McColl EM, Wheeler GD, Gomes P, Bhambhani Y, Cumming DC. The effects of acute exercise on pulsatile LH release in high-mileage male runners. Clin Endocrinol. 1989;31(5):617–21. 93. Hackney AC, Sinning WE, Bruot BC. Reproductive hormonal profiles of endurance-trained and untrained males. Med Sci Sports Exerc. 1988;20(1):60–5. 94. Arce JC, De Souza MJ, Pescatello LS, Luciano AA. Subclinical alterations in hormone and semen profile in athletes. Fertil Steril. 1993;59(2):398–404. 95. Hackney AC, Fahrner CL, Gulledge TP. Basal reproductive hormonal profiles are altered in endurance trained men. J Sports Med Phys Fitness. 1998;38(2):138–41. 96. Hackney AC, Sinning WE, Bruot BC. Hypothalamic-­ pituitary-­testicular axis function in endurance-trained males. Int J Sports Med. 1990;11(4):298–303. 97. Roberts AC, McClure RD, Weiner RI, Brooks GA. Overtraining affects male reproductive status. Fertil Steril. 1993;60(4):686–92. 98. Wheeler GD, Singh M, Pierce WD, Epling WF, Cumming DC. Endurance training decreases serum testosterone levels in men without change in luteinizing hormone pulsatile release. J Clin Endocrinol Metab. 1991;72(2):422–5. 99. Griffith RO, Dressendorfer RH, Fullbright CD, Wade CE. Testicular function during exhaustive training. Phys Sports Med. 1990;18:54–64. 100. Vasankari TJ, Kujala UM, Heinonen OJ, Huhtaniemi IT. Effects of endurance training on hormonal responses to prolonged physical exercise in males. Acta Endocrinol. 1993;129(2):109–13. 101. Hackney AC, Sharp RL, Runyan WS, Ness RJ. Relationship of resting prolactin and testosterone in males during intensive training. Br J Sports Med. 1989;23(3):194. 102. Urhausen A, Kullmer T, Kindermann W. A 7-week follow-up study of the behaviour of testosterone and 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. 116. 243 cortisol during the competition period in rowers. Eur J Appl Physiol. 1987;56(5):528–33. Maïmoun L, Lumbroso S, Manetta J, Paris F, Leroux JL, Sultan C. Testosterone is significantly reduced in endurance athletes without impact on bone mineral density. Horm Res Basel. 2003;59(6):285–92. Duplanty AA, Levitt DE, Hill DW, McFarlin BK, DiMarco NM, Vingren JL. Resistance training is associated with higher bone mineral density among young adult male distance runners independent of physiological factors. J Strength Cond Res. 2018;32(6):1594–600. MacConnie SE, Barkan A, Lampman RM, Schork MA, Beitins IZ. Decreased hypothalamic gonadotropin-­ releasing hormone secretion in male marathon runners. N Engl J Med. 1986;315(7):411–7. Keay N, Francis G, Hind K. Low energy availability assessed by a sport-specific questionnaire and clinical interview indicative of bone health, endocrine profile and cycling performance in competitive male cyclists. BMJ Open Sport Exerc Med. 2018;4(1):e000424. Hetland ML, Haarbo J, Christiansen C. Low bone mass and high bone turnover in male long distance runners. J Clin Endocrinol Metab. 1993;77(3):770–5. Fredericson M, Chew K, Ngo J, Cleek T, Kiratli J, Cobb K. Regional bone mineral density in male athletes: a comparison of soccer players, runners and controls. Br J Sports Med. 2007;41(10):664–8. discussion 668. Hind K, Truscott JG, Evans JA. Low lumbar spine bone mineral density in both male and female endurance runners. Bone. 2006;39(4):880–5. MacDougall JD, Webber CE, Martin J, Ormerod S, Chesley A, Younglai EV, et al. Relationship among running mileage, bone density, and serum testosterone in male runners. J Appl Physiol 1985. 1992;73(3):1165–70. Bilanin JE, Blanchard MS, Russek-Cohen E. Lower vertebral bone density in male long distance runners. Med Sci Sports Exerc. 1989;21(1):66–70. Smathers AM, Bemben MG, Bemben DA. Bone density comparisons in male competitive road cyclists and untrained controls. Med Sci Sports Exerc. 2009;41(2):290–6. Nichols JF, Rauh MJ. Longitudinal changes in bone mineral density in male master cyclists and nonathletes. J Strength Cond Res. 2011;25(3):727–34. Stewart AD, Hannan J. Total and regional bone density in male runners, cyclists, and controls. Med Sci Sports Exerc. 2000;32(8):1373–7. MacKelvie KJ, Taunton JE, McKay HA, Khan KM. Bone mineral density and serum testosterone in chronically trained, high mileage 40–55 year old male runners. Br J Sports Med. 2000;34(4):273–8. Burge MR, Lanzi RA, Skarda ST, Eaton RP. Idiopathic hypogonadotropic hypogonadism in a male runner is reversed by clomiphene citrate. Fertil Steril. 1997;67(4):783–5. W. R.D. Duff and P. D. Chilibeck 244 117. Remes T, Väisänen SB, Mahonen A, Huuskonen J, Kröger H, Jurvelin JS, et al. The association of bone metabolism with bone mineral density, serum sex hormone concentrations, and regular exercise in middle-aged men. Bone. 2004;35(2):439–47. 118. Maïmoun L, Sultan C. Effect of physical activity on calcium homeostasis and calciotropic hormones: a review. Calcif Tissue Int. 2009;85(4):277–86. 119. McSheehy PM, Chambers TJ. Osteoblast-like cells in the presence of parathyroid hormone release soluble factor that stimulates osteoclastic bone resorption. Endocrinology. 1986;119(4):1654–9. 120. Bauer W, Aub JC, Albright F. Studies of calcium and phosphorous metabolism: v. a study of the bone trabeculae as a readily available reserve supply of calcium. J Exp Med. 1929;49(1):145–62. 121. Scott JPR, Sale C, Greeves JP, Casey A, Dutton J, Fraser WD. Treadmill running reduces parathyroid hormone concentrations during recovery compared with a nonexercising control group. J Clin Endocrinol Metab. 2014;99(5):1774–82. 122. Frolik CA, Black EC, Cain RL, Satterwhite JH, Brown-Augsburger PL, Sato M, et al. Anabolic and catabolic bone effects of human parathyroid hormone (1-34) are predicted by duration of hormone exposure. Bone. 2003;33(3):372–9. 123. Bouassida A, Latiri I, Bouassida S, Zalleg D, Zaouali M, Feki Y, et al. Parathyroid hormone and physical exercise: a brief review. J Sports Sci Med. 2006;5(3):367–74. 124. Dempster DW, Cosman F, Parisien M, Shen V, Lindsay R. Anabolic actions of parathyroid hormone on bone. Endocr Rev. 1993;14(6):690–709. 125. Gardinier JD, Mohamed F, Kohn DH. PTH signaling during exercise contributes to bone adaptation. J Bone Miner Res. 2015;30(6):1053–63. 126. Ljunghall S, Joborn H, Roxin LE, Skarfors ET, Wide LE, Lithell HO. Increase in serum parathyroid hormone levels after prolonged physical exercise. Med Sci Sports Exerc. 1988;20(2):122–5. 127. Barry DW, Kohrt WM. Acute effects of 2 hours of moderate-intensity cycling on serum parathyroid hormone and calcium. Calcif Tissue Int. 2007;80(6):359–65. 128. Maïmoun L, Simar D, Malatesta D, Caillaud C, Peruchon E, Couret I, et al. Response of bone metabolism related hormones to a single session of strenuous exercise in active elderly subjects. Br J Sports Med. 2005;39(8):497–502. 129. Bouassida A, Zalleg D, Zaouali Ajina M, Gharbi N, Duclos M, Richalet JP, et al. Parathyroid hormone concentrations during and after two periods of high intensity exercise with and without an intervening recovery period. Eur J Appl Physiol. 2003;88(4–5):339–44. 130. Brahm H, Ström H, Piehl-Aulin K, Mallmin H, Ljunghall S. Bone metabolism in endurance trained athletes: a comparison to population-based controls based on DXA, SXA, quantitative ultra- 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. sound, and biochemical markers. Calcif Tissue Int. 1997;61(6):448–54. Thorsen K, Kristoffersson A, Hultdin J, Lorentzon R. Effects of moderate endurance exercise on calcium, parathyroid hormone, and markers of bone metabolism in young women. Calcif Tissue Int. 1997;60(1):16–20. Tosun A, Bölükbaşi N, Cingi E, Beyazova M, Unlü M. Acute effects of a single session of aerobic exercise with or without weight-lifting on bone turnover in healthy young women. Mod Rheumatol. 2006;16(5):300–4. Aloia JF, Rasulo P, Deftos LJ, Vaswani A, Yeh JK. Exercise-induced hypercalcemia and the calciotropic hormones. J Lab Clin Med. 1985;106(3):229–32. Grimston SK, Tanguay KE, Gundberg CM, Hanley DA. The calciotropic hormone response to changes in serum calcium during exercise in female long distance runners. J Clin Endocrinol Metab. 1993;76(4):867–72. Brahm H, Piehl-Aulin K, Saltin B, Ljunghall S. Net fluxes over working thigh of hormones, growth factors and biomarkers of bone metabolism during short lasting dynamic exercise. Calcif Tissue Int. 1997;60(2):175–80. Kristoffersson A, Hultdin J, Holmlund I, Thorsen K, Lorentzon R. Effects of short-term maximal work on plasma calcium, parathyroid hormone, osteocalcin and biochemical markers of collagen metabolism. Int J Sports Med. 1995;16(3):145–9. Maimoun L, Manetta J, Couret I, Dupuy AM, Mariano-Goulart D, Micallef JP, et al. The intensity level of physical exercise and the bone metabolism response. J Sports Med. 2006;27(2):105–11. Ryder KD, Duncan RL. Parathyroid hormone modulates the response of osteoblast-like cells to mechanical stimulation. Calcif Tissue Int. 2000;67(3):241–6. Scott JPR, Sale C, Greeves JP, Casey A, Dutton J, Fraser WD. The effect of training status on the metabolic response of bone to an acute bout of exhaustive treadmill running. J Clin Endocrinol Metab. 2010;95(8):3918–25. Kohrt WM, Wherry SJ, Wolfe P, Sherk VD, Wellington T, Swanson CM, et al. Maintenance of serum ionized calcium during exercise attenuates parathyroid hormone and bone resorption responses. J Bone Miner Res. 2018;33(7):1326–34. Barry DW, Hansen KC, van Pelt RE, Witten M, Wolfe P, Kohrt WM. Acute calcium ingestion attenuates exercise-induced disruption of calcium homeostasis. Med Sci Sports Exerc. 2011;43(4):617–23. Zerath E, Holy X, Douce P, Guezennec CY, Chatard JC. Effect of endurance training on postexercise parathyroid hormone levels in elderly men. Med Sci Sports Exerc. 1997;29(9):1139–45. Lester ME, Urso ML, Evans RK, Pierce JR, Spiering BA, Maresh CM, et al. Influence of exercise mode and osteogenic index on bone biomarker 14 144. 145. 146. 147. 148. 149. 150. 151. 152. 153. 154. 155. 156. 157. Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone responses during short-term physical training. Bone. 2009;45(4):768–76. Maïmoun L, Galy O, Manetta J, Coste O, Peruchon E, Micallef JP, et al. Competitive season of triathlon does not alter bone metabolism and bone ­mineral status in male triathletes. Int J Sports Med. 2004;25(3):230–4. Brown EM, Hurwitz S, Aurbach GD. Beta-adrenergic stimulation of cyclic AMP content and parathyroid hormone release from isolated bovine parathyroid cells. Endocrinology. 1977;100(6):1696–702. Salvesen H, Johansson AG, Foxdal P, Wide L, Piehl-Aulin K, Ljunghall S. Intact serum parathyroid hormone levels increase during running exercise in well-trained men. Calcif Tissue Int. 1994;54(4):256–61. Rockwell JC, Sorensen AM, Baker S, Leahey D, Stock JL, Michaels J, et al. Weight training decreases vertebral bone density in premenopausal women: a prospective study. J Clin Endocrinol Metab. 1990;71(4):988–93. Bloomfield SA, Mysiw WJ, Jackson RD. Bone mass and endocrine adaptations to training in spinal cord injured individuals. Bone. 1996;19(1):61–8. Nelson ME, Fiatarone MA, Morganti CM, Trice I, Greenberg RA, Evans WJ. Effects of high-intensity strength training on multiple risk factors for osteoporotic fractures. A randomized controlled trial. JAMA. 1994;272(24):1909–14. Boucher A, D’Amour P, Hamel L, Fugère P, Gascon-­ Barré M, Lepage R, et al. Estrogen replacement decreases the set point of parathyroid hormone stimulation by calcium in normal postmenopausal women. J Clin Endocrinol Metab. 1989;68(4):831–6. Owens DJ, Allison R, Close GL. Vitamin D and the athlete: current perspectives and new challenges. Sports Med Auckl NZ. 2018;48(Suppl 1):3–16. Chen TC, Chimeh F, Lu Z, Mathieu J, Person KS, Zhang A, et al. Factors that influence the cutaneous synthesis and dietary sources of vitamin D. Arch Biochem Biophys. 2007;460(2):213–7. Rector RS, Rogers R, Ruebel M, Hinton PS. Participation in road cycling vs running is associated with lower bone mineral density in men. Metabolism. 2008;57(2):226–32. Backx E, van der Avoort C, Tieland M, Maase K, Kies A, van Loon L, et al. Seasonal variation in vitamin D status in elite athletes: a longitudinal study. Int J Sport Nutr Exerc Metab. 2017;27(1):6–10. Bescós García R, Rodríguez Guisado FA. Low levels of vitamin D in professional basketball players after wintertime: relationship with dietary intake of vitamin D and calcium. Nutr Hosp. 2011;26(5):945–51. Fishman MP, Lombardo SJ, Kharrazi FD. Vitamin D deficiency among professional basketball players. Orthop J Sports Med. 2016;4(7):2325967116655742. Hamilton B, Grantham J, Racinais S, Chalabi H. Vitamin D deficiency is endemic in Middle Eastern sportsmen. Public Health Nutr. 2010;13(10):1528–34. 245 158. Krzywanski J, Mikulski T, Krysztofiak H, Mlynczak M, Gaczynska E, Ziemba A. Seasonal vitamin D status in polish elite athletes in relation to sun exposure and oral supplementation. PLoS One. 2016;11(10):e0164395. 159. Magee PJ, Pourshahidi LK, Wallace JMW, Cleary J, Conway J, Harney E, et al. Vitamin D status and supplementation in elite Irish athletes. Int J Sport Nutr Exerc Metab. 2013;23(5):441–8. 160. Morton JP, Iqbal Z, Drust B, Burgess D, Close GL, Brukner PD. Seasonal variation in vitamin D status in professional soccer players of the English Premier League. Appl Physiol Nutr Metab. 2012;37(4):798–802. 161. Sghaier-Ayadi A, Feki M, Ayed IB, Abene O, Fredj MB, Kaabachi K, et al. Vitamin D status and determinants of deficiency in non-supplemented athletes during the winter months in Tunisia. Biol Sport. 2015;32(4):281–7. 162. Valtueña J, Dominguez D, Til L, González-Gross M, Drobnic F. High prevalence of vitamin D insufficiency among elite Spanish athletes the importance of outdoor training adaptation. Nutr Hosp. 2014;30(1):124–31. 163. Allison RJ, Farooq A, Hamilton B, Close GL, Wilson MG. No association between vitamin D deficiency and markers of bone health in athletes. Med Sci Sports Exerc. 2015;47(4):782–8. 164. Ludwa IA, Falk B, Yao M, Corbett L, Klentrou P. Bone speed of sound, bone turnover and IGF-I in adolescent synchronized swimmers. Pediatr Exerc Sci. 2010;22(3):421–30. 165. Yeh JK, Liu CC, Aloia JF. Additive effect of treadmill exercise and 17 beta-estradiol replacement on prevention of tibial bone loss in adult ovariectomized rat. J Bone Miner Res. 1993;8(6):677–83. 166. Cheng MZ, Zaman G, Rawlinson SC, Suswillo RF, Lanyon LE. Mechanical loading and sex hormone interactions in organ cultures of rat ulna. J Bone Miner Res Off J Am Soc Bone Miner Res. 1996;11(4):502–11. 167. Souza MVC, Lino ADS, Ruffoni LGD, Domingos MM, Barbosa MR, Rodrigues MFC, et al. Resistance training and hormone replacement increase MMP-2 activity, quality and quantity of bone in ovariectomized rats. Mot Rev Educ Física [Internet]. 2017 [cited 2019 Jan 7];23(4). Available from: http://www.scielo.br/scielo.php?script=sci_ abstract&pid=S1980-65742017000400303&lng=en &nrm=iso&tlng=en. 168. Kohrt WM, Snead DB, Slatopolsky E, Birge SJ. Additive effects of weight-bearing exercise and estrogen on bone mineral density in older women. J Bone Miner Res Off J Am Soc Bone Miner Res. 1995;10(9):1303–11. 169. Villareal DT, Binder EF, Yarasheski KE, Williams DB, Brown M, Sinacore DR, et al. Effects of exercise training added to ongoing hormone replacement therapy on bone mineral density in frail elderly women. J Am Geriatr Soc. 2003;51(7):985–90. W. R.D. Duff and P. D. Chilibeck 246 170. Notelovitz M, Martin D, Tesar R, Khan FY, Probart C, Fields C, et al. Estrogen therapy and variable-­ resistance weight training increase bone mineral in surgically menopausal women. J Bone Miner Res Off J Am Soc Bone Miner Res. 1991;6(6):583–90. 171. Milliken LA, Going SB, Houtkooper LB, Flint-­ Wagner HG, Figueroa A, Metcalfe LL, et al. Effects of exercise training on bone remodeling, insulin-like growth factors, and bone mineral density in postmenopausal women with and without hormone replacement therapy. Calcif Tissue Int. 2003;72(4):478–84. 172. Maddalozzo GF, Widrick JJ, Cardinal BJ, Winters-­ Stone KM, Hoffman MA, Snow CM. The effects of hormone replacement therapy and resistance training on spine bone mineral density in early postmenopausal women. Bone. 2007;40(5):1244–51. 173. Zhao R, Xu Z, Zhao M. Antiresorptive agents increase the effects of exercise on preventing postmenopausal bone loss in women: a meta-analysis. PLoS One. 2015;10(1):e0116729. 174. Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the women’s health initiative randomized controlled trial. JAMA. 2002;288(3):321–33. 175. Nakajima D, Kim CS, Oh TW, Yang CY, Naka T, Igawa S, et al. Suppressive effects of genistein dosage and resistance exercise on bone loss in ovariectomized rats. J Physiol Anthropol Appl Hum Sci. 2001;20(5):285–91. 176. Wu J, Wang XX, Takasaki M, Ohta A, Higuchi M, Ishimi Y. Cooperative effects of exercise training and genistein administration on bone mass in ovariectomized mice. J Bone Miner Res Off J Am Soc Bone Miner Res. 2001;16(10):1829–36. 177. Wu J, Wang X, Chiba H, Higuchi M, Nakatani T, Ezaki O, et al. Combined intervention of soy isoflavone and moderate exercise prevents body fat elevation and bone loss in ovariectomized mice. Metabolism. 2004;53(7):942–8. 178. Chilibeck PD, Vatanparast H, Pierson R, Case A, Olatunbosun O, Whiting SJ, et al. Effect of exercise training combined with isoflavone supplementation on bone and lipids in postmenopausal women: a randomized clinical trial. J Bone Miner Res. 2013;28(4):780–93. 179. Saxon LK, Turner CH. Estrogen receptor beta: the antimechanostat? Bone. 2005;36(2):185–92. 180. Mathews LS, Norstedt G, Palmiter RD. Regulation of insulin-like growth factor I gene expression by growth hormone. Proc Natl Acad Sci U S A. 1986;83(24):9343–7. 181. Kassem M, Blum W, Ristelli J, Mosekilde L, Eriksen EF. Growth hormone stimulates proliferation and differentiation of normal human osteoblast-like cells in vitro. Calcif Tissue Int. 1993;52(3):222–6. 182. Schmid C, Guler HP, Rowe D, Froesch ER. Insulin-­ like growth factor I regulates type I procollagen mes- 183. 184. 185. 186. 187. 188. 189. 190. 191. 192. 193. 194. 195. 196. 197. senger ribonucleic acid steady state levels in bone of rats. Endocrinology. 1989;125(3):1575–80. Kraemer RR, Kilgore JL, Kraemer GR, Castracane VD. Growth hormone, IGF-I, and testosterone responses to resistive exercise. Med Sci Sports Exerc. 1992;24(12):1346–52. Häkkinen K, Pakarinen A. Acute hormonal responses to two different fatiguing heavy-resistance protocols in male athletes. J Appl Physiol 1985. 1993;74(2):882–7. Craig BW, Brown R, Everhart J. Effects of progressive resistance training on growth hormone and testosterone levels in young and elderly subjects. Mech Ageing Dev. 1989;49(2):159–69. Cappon J, Brasel JA, Mohan S. Cooper DM. Effect of brief exercise on circulating insulin-like growth factor I. J Appl Physiol 1985. 1994;76(6):2490–6. Kraemer WJ, Marchitelli L, Gordon SE, Harman E, Dziados JE, Mello R, et al. Hormonal and growth factor responses to heavy resistance exercise protocols. J Appl Physiol 1985. 1990;69(4):1442–50. Cumming DC, Brunsting LA, Strich G, Ries AL, Rebar RW. Reproductive hormone increases in response to acute exercise in men. Med Sci Sports Exerc. 1986;18(4):369–73. Fahrner CL, Hackney AC. Effects of endurance exercise on free testosterone concentration and the binding affinity of sex hormone binding globulin (SHBG). Int J Sports Med. 1998;19(1):12–5. Kraemer RR, Heleniak RJ, Tryniecki JL, Kraemer GR, Okazaki NJ, Castracane VD. Follicular and luteal phase hormonal responses to low-­ volume resistive exercise. Med Sci Sports Exerc. 1995;27(6):809–17. Copeland JL, Consitt LA, Tremblay MS. Hormonal responses to endurance and resistance exercise in females aged 19-69 years. J Gerontol A Biol Sci Med Sci. 2002;57(4):B158–65. Chadan SG, Dill RP, Vanderhoek K, Parkhouse WS. Influence of physical activity on plasma insulin-­ like growth factor-1 and insulin-like growth factor binding proteins in healthy older women. Mech Ageing Dev. 1999;109(1):21–34. Kraemer WJ, Gordon SE, Fleck SJ, Marchitelli LJ, Mello R, Dziados JE, et al. Endogenous anabolic hormonal and growth factor responses to heavy resistance exercise in males and females. Int J Sports Med. 1991;12(2):228–35. Cumming DC, Wall SR, Galbraith MA, Belcastro AN. Reproductive hormone responses to resistance exercise. Med Sci Sports Exerc. 1987;19(3):234–8. Godfrey RJ, Madgwick Z, Whyte GP. The exercise-­ induced growth hormone response in athletes. Sports Med Auckl NZ. 2003;33(8):599–613. Kraemer WJ, Ratamess NA. Hormonal responses and adaptations to resistance exercise and training. Sports Med Auckl NZ. 2005;35(4):339–61. Storey A, Smith HK. Unique aspects of competitive weightlifting: performance, training and physiology. Sports Med Auckl NZ. 2012;42(9):769–90. 14 Hormonal Regulation of the Positive and Negative Effects of Exercise on Bone 198. Häkkinen K, Pakarinen A. Acute hormonal responses to heavy resistance exercise in men and women at different ages. Int J Sports Med. 1995;16(8):507–13. 199. Häkkinen K, Pakarinen A, Kraemer WJ, Newton RU, Alen M. Basal concentrations and acute responses of serum hormones and strength development during heavy resistance training in middle-aged and elderly men and women. J Gerontol A Biol Sci Med Sci. 2000;55(2):B95–105. 200. Copeland JL. Anabolic hormones in aging women: effects of supplementation vs. physical activity. Can J Appl Physiol. 2004;29(1):76–89. 201. Wallace JD, Cuneo RC, Baxter R, Orskov H, Keay N, Pentecost C, et al. Responses of the growth hormone (GH) and insulin-like growth factor axis to exercise, GH administration, and GH withdrawal in trained adult males: a potential test for GH abuse in sport. J Clin Endocrinol Metab. 1999;84(10):3591–601. 202. Wallace JD, Cuneo RC, Lundberg PA, Rosen T, Jorgensen JO, Longobardi S, et al. Responses of markers of bone and collagen turnover to exercise, growth hormone (GH) administration, and GH withdrawal in trained adult males. J Clin Endocrinol. 2000;85(1):124–33. 203. Tsuchiya Y, Sakuraba K, Ochi E. High force eccentric exercise enhances serum tartrate-resistant acid phosphatase-5b and osteocalcin. J Musculoskelet Neuronal Interact. 2014;14(1):50–7. 204. Davee AM, Rosen CJ, Adler RA. Exercise patterns and trabecular bone density in college women. J Bone Miner Res Off J Am Soc Bone Miner Res. 1990;5(3):245–50. 205. Snow CM, Rosen CJ, Robinson TL. Serum IGF-I is higher in gymnasts than runners and predicts bone and lean mass. Med Sci Sports Exerc. 2000;32(11):1902–7. 206. Buchanan JR, Myers C, Lloyd T, Leuenberger P, Demers LM. Determinants of peak trabecular bone density in women: the role of androgens, estrogen, and exercise. J Bone Miner Res. 1988;3(6):673–80. 207. Nelson ME, Meredith CN, Dawson-Hughes B, Evans WJ. Hormone and bone mineral status in endurance-­ trained and sedentary postmenopausal women. J Clin Endocrinol Metab. 1988;66(5):927–33. 208. Nowak A, Straburzyńska-Lupa A, Kusy K, Zieliński J, Felsenberg D, Rittweger J, et al. Bone mineral density and bone turnover in male masters athletes aged 40-64. Aging Male. 2010;13(2):133–41. 209. Ay A, Yurtkuran M. Evaluation of hormonal response and ultrasonic changes in the heel bone by aquatic exercise in sedentary postmenopausal women. Am J Phys Med Rehabil. 2003;82(12):942–9. 210. Hinton PS, Nigh P, Thyfault J. Serum sclerostin decreases following 12months of resistance- or jump-training in men with low bone mass. Bone. 2017;96:85–90. 211. Duff WRD, Chilibeck PD, Rooke JJ, Kaviani M, Krentz JR, Haines DM. The effect of bovine colostrum supplementation in older adults during 212. 213. 214. 215. 216. 217. 218. 219. 220. 221. 222. 223. 224. 225. 247 resistance training. Int J Sport Nutr Exerc Metab. 2014;24(3):276–85. Ryan AS, Treuth MS, Rubin MA, Miller JP, Nicklas BJ, Landis DM, et al. Effects of strength training on bone mineral density: hormonal and bone turnover relationships. J Appl Physiol Bethesda Md 1985. 1994;77(4):1678–84. Maddalozzo GF, Snow CM. High intensity resistance training: effects on bone in older men and women. Calcif Tissue Int. 2000;66(6):399–404. Nichols DL, Sanborn CF, Bonnick SL, Ben-Ezra V, Gench B, DiMarco NM. The effects of gymnastics training on bone mineral density. Med Sci Sports Exerc. 1994;26(10):1220–5. Daly RM, Rich PA, Klein R, Bass S. Effects of high-­ impact exercise on ultrasonic and biochemical indices of skeletal status: a prospective study in young male gymnasts. J Bone. 1999;14(7):1222–30. Henderson SA, Graham HK, Mollan RAB, Riddoch C, Sheridan B, Johnston H. Calcium homeostasis and exercise. Int Orthop. 1989;13(1):69–73. Rong H, Berg U, Tørring O, Sundberg CJ, Granberg B, Bucht E. Effect of acute endurance and strength exercise on circulating calcium-regulating hormones and bone markers in young healthy males. Scand J Med Sci Sports. 1997;7(3):152–9. Yeh JK, Aloia JF. Effect of physical activity on calciotropic hormones and calcium balance in rats. Am J Phys. 1990;258(2 Pt 1):E263–8. Smith JK, Dykes R, Chi DS. The effect of long-term exercise on the production of osteoclastogenic and antiosteoclastogenic cytokines by peripheral blood mononuclear cells and on serum markers of bone metabolism [Internet]. J Osteoporos. 2016 [cited 2019 Jan 6]. Available from: https://www.hindawi. com/journals/jos/2016/5925380/. Fujimura R, Ashizawa N, Watanabe M, Mukai N, Amagai H, Fukubayashi T, et al. Effect of resistance exercise training on bone formation and resorption in young male subjects assessed by biomarkers of bone metabolism. J Bone Miner Res. 1997;12(4):656–62. Bell NH, Godsen RN, Henry DP, Shary J, Epstein S. The effects of muscle-building exercise on vitamin D and mineral metabolism. J Bone Miner Res. 1988;3(4):369–73. Maïmoun L, Coste O, Puech A-M, Peruchon E, Jaussent A, Paris F, et al. No negative impact of reduced leptin secretion on bone metabolism in male decathletes. Eur J Appl Physiol. 2007;102(3):343–51. Zittermann A, Sabatschus O, Jantzen S, Platen P, Danz A, Dimitriou T, et al. Exercise-trained young men have higher calcium absorption rates and plasma calcitriol levels compared with age-matched sedentary controls. Calcif Tissue Int. 2000;67(3):215–9. Yeh JK, Aloia JF, Yasumura S. Effect of physical activity on calcium and phosphorus metabolism in the rat. Am J Phys. 1989;256(1 Pt 1):E1–6. Spanos E, Barrett D, Macintyre I, Pike JW, Safilian EF, Haussler MR. Effect of growth hormone on vitamin D metabolism. Nature. 1978;273(5659):246–7. Interrelations Between Acute and Chronic Exercise Stress and the Immune and Endocrine Systems 15 Jonathan Peake Introduction Interaction between the endocrine and immune system is necessary to regulate our health. However, under some conditions, stress hormones can overstimulate or suppress the immune system, resulting in harmful consequences [1]. Stress is often considered negative, yet it is an intrinsic part of everyday life. Stress is not clearly defined; it is context-specific and depends on the nature of factors that challenge our body. Internal stimuli will elicit different stress reactions compared with external stimuli [1]. Similarly, some stressors will induce responses that may benefit survival, whereas others will cause disturbances that may endanger our health. Stress also depends on how our bodies perceive and respond to stressful stimuli [1]. Several important factors determine whether stress hormones stimulate or inhibit the immune system. These factors include [1]: • The effects of stress on the distribution of immune cells in the body • The duration of stress • Hormone concentrations • The timing of stress hormone exposure relative to the activation status of immune cells (i.e., naїve vs. activated, early vs. late activation) J. Peake (*) School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia e-mail: jonathan.peake@qut.edu.au Exercise is a reproducible and quantifiable model of stress and is useful for studying the interactions between the endocrine and immune systems. Exercise stimulates the secretion of a variety of stress hormones, but catecholamines, cortisol and growth hormone are most closely linked with exercise-induced changes in immune function. Research on the interactions between endocrine and immune systems following acute exercise and chronic training is important. Regular exposure to mild short-term stress can potentially enhance immune function and lead to various health benefits. Conversely, prolonged exposure to the chronic stress of intense training may inhibit certain immune functions that are required for health maintenance. This chapter describes the regulatory roles of stress hormones on immune cell counts and activity during acute exercise and following chronic exercise training. Figure 15.1 summarises the immunoendocrine interactions during exercise and their potential functional significance. echanisms of Interaction: In Vitro M Evidence Stress hormones modulate immune function directly by binding to cognate receptors on immune cells and indirectly by modulating the production of cytokines (e.g., IFN-γ, IL-1β, IL-6, TNF-α) [2]. Glucocorticoid receptors are © Springer Nature Switzerland AG 2020 A. C. Hackney, N. W. Constantini (eds.), Endocrinology of Physical Activity and Sport, Contemporary Endocrinology, https://doi.org/10.1007/978-3-030-33376-8_15 249 J. Peake 250 Brain Exercise Sympathetic nerve fibre Heart Neuroendocrine Skin HPA axis Lung mucosa Adrenal gland ↑ Cardiac output ↑ Shear stress Demargination from vascular pools -Spleen? -Lung? -Liver? -Active muscles? Peripheral circulation ACTH Gut Medulla Cortex Catecholamines Cortisol Cell trafficking -Adhesion molecules -Apoptosis Other immune mediators -Cytokines -Chemokines -Heat shock proteins Preferential mobilisation of cells with altered effector phenotype? Effector functions -Microbial killing -Cytokine expression Tissue migration/ homing Fig. 15.1 Potential mechanisms by which stress hormone interacts with the immune system during exercise. (Modified from TOM-Systemdruck GmbH, Walsh et al. [170]) expressed on monocytes and B lymphocytes, whereas glucocorticoid receptor expression is much lower on CD3+ T cells and neutrophils [3, 4]. β2-adrenoreceptors for catecholamines are expressed on (in descending order) natural killer (NK) cells, monocytes, B lymphocytes and T suppressor lymphocytes [5]. Macrophages [6] and neutrophils [7] also express β2-­adrenoreceptors. Within T lymphocyte subpopulations, β2adrenoreceptors are mainly expressed in naїve CD4+ T cells and T helper 1 and T helper 2 cells [8–10]. mRNA for α-adrenoreceptors is expressed by activated T cells [11] and in peripheral blood mononuclear cells of patients with juvenile rheumatoid arthritis but not healthy individuals [12]. Although B lymphocytes, monocytes and neutrophils all express growth hormone receptors [13–15], growth hormone most likely exerts its effects on the immune system by binding to prolactin receptors, which are expressed on monocytes and B and T cells [16]. Immune cells also express receptors for other stress hormones, including substance P [17], neuropeptide Y [18], corticotrophin-releasing hormone [19] and serotonin [20]. Glucocorticoids regulate the activity of immune cells by binding to glucocorticoid receptors, which in turn suppresses the transcription factors activator protein 1 (AP-1) and nuclear factor κ B (NFκB) [21]. Glucocorticoids inhibit AP-1 transcriptional activity by preventing the oncoproteins c-Fos and c-Jun from binding to the AP-1 consensus binding site in DNA [22]. Glucocorticoids inhibit NFκB transcriptional activity through two mechanisms. 15 Interrelations Between Acute and Chronic Exercise Stress and the Immune and Endocrine Systems Firstly, glucocorticoids can induce expression of the inhibitory protein IκB, which then prevents NFκB from translocating to the nucleus where it initiates transcription [23]. Secondly, physical interaction or cross-talk between glucocorticoid receptors and NFκB can suppress transcription [24, 25]. By suppressing the transcriptional activity of AP-1 and NFκB, glucocorticoids regulate various immune functions, including cytokine production [21]. In particular, glucocorticoids inhibit monocyte production of type 1 cytokines IL-12 and IFN-γ, which in turn favours the production of type 2 cytokines IL-4 and IL-10 by CD4+ lymphocytes and peripheral blood mononuclear cells [26–29]. Type 1 cytokines regulate the activity T cytotoxic cells, NK cells and macrophages which defend against intracellular pathogens. Type 2 cytokines regulate the activity of B lymphocytes, eosinophils and mast cells, which defend against extracellular pathogens [30]. The type 1/type 2 cytokine balance determines the balance between cell-mediated vs. humoral immunity and the risk of various immune-related disorders [31]. For information on the effects of glucocorticoids on other aspects of immune function, readers are referred to other more comprehensive reviews [21, 32]. Binding of catecholamines to β2-­ adrenoreceptors can inhibit IL-2 and IFN-γ and stimulate IL-4 and IL-10 production by T cells and peripheral blood mononuclear cells [26, 33, 34]. Similar to glucocorticoids, catecholamines can therefore induce a shift towards type 2 cytokine production. The combined effects of glucocorticoids and catecholamines on IFN-γ, IL-4 and IL-10 production by peripheral blood mononuclear cells are in fact additive [26]. However, there are some inconsistencies in the literature concerning the effects of β-agonists on cytokine production. Some studies report that T helper 2 lymphocytes do not respond to β-agonist stimulation [9, 35], but more recent data indicate that activated T cells do produce cytokines following β-agonist stimulation [10]. The effects of β-agonists on cytokine production may also be dose-dependent. Low concentrations of β-agonists (i.e., 1–10 nM) stimulate cytokine production, whereas high concentrations (i.e., 100 nM to 251 10 μM) inhibit cytokine production by T cells [10]. Downstream from cyclic AMP, β-agonists inhibit cytokine production by T cells by blocking the calcium-/calmodulin-dependent protein phosphatase calcineurin and p38 mitogen-­ activated protein kinase, but not NFκB [10, 36]. For information on the effects of catecholamines on other aspects of immune function, readers are referred to other more comprehensive reviews [21, 31, 37]. In comparison with glucocorticoids and catecholamines, less is known about the effects of growth hormone and prolactin on the immune system. The actions of growth hormone and insulin-­like growth factor-1 (IGF-1) do not overlap entirely, but growth hormone exerts many of its actions through IGF-1. Neither growth hormone nor IGF-1 is essential for immune function, but growth hormone influences various aspects of immune cell development and activity [38]. Growth hormone inhibits apoptosis of CD4+ T cells following treatment with dexamethasone [39]. Growth hormone, through binding to its receptor on the surface of T cells, may activate phosphatidylinositol 3 kinase (which regulates cell proliferation) and NFκB (which controls apoptosis through the anti-apoptosis protein Bcl2) [40]. IGF-1 also stimulates macrophages to produce reactive oxygen species [41] and increases NK cell activity [42]. Prolactin is also not essential to normal immune function [38], but it can promote lymphocyte proliferation [43] and haematopoiesis [44]. Interactions between the neuroendocrine and immune systems are bidirectional. Pro-­ inflammatory cytokines released from immune cells (e.g., IL-1β, IL-6 and TNF-α) mediate communication between the immune system and the central nervous system. Cytokines can alter activity of the central nervous system through humoral, neural and cellular pathways [45]. Cytokines can pass the blood–brain barrier directly [46]. Alternatively, immune cells can pass across the blood–brain barrier and release cytokines into the central nervous system [47]. Cells comprising the blood–brain barrier also secrete various cytokines [48]. Cytokines may signal the central nervous system by stimulating afferent nerves, although this concept remains J. Peake 252 somewhat controversial [49]. One theory proposes that cytokines target the blood–brain barrier during systemic inflammation, whereas they target afferent nerves during localised inflammation [49]. Cytokines can pass back across the blood–brain barrier into the circulation following intracerebroventricular injection of lipopolysaccharide (LPS) [50]. Cytokines interact with components of the central nervous system, resulting in behavioural changes. Specifically, cytokines alter neurotransmitter function, neuroendocrine activity, neural plasticity and neural circuitry. These actions can induce fever, changes in appetite, fatigue and depression [45]. tress Hormones and Leukocyte S Mobilisation In Vivo A number of studies have investigated the effects of stress hormones on circulating leukocyte numbers by infusing variable doses of stress hormones in healthy humans over 30 min up to 5 h. Cortisol raises the number of circulating neutrophils, whereas it suppresses the number of lymphocytes, and does not alter the number of Leu+ NK cells [51, 52]. By contrast, adrenaline increases the number of circulating total lymphocytes and NK cells [51, 53–55]. The number of circulating monocytes also rises 1–2 h following infusion of adrenaline [53, 55, 56]. In contrast with NK cells, the effects of adrenaline and the β-agonist isoproterenol on circulating T lymphocyte subpopulations are somewhat variable. In response to these agents, the number/percentage of circulating CD4+ T helper cells decreases [54, 56, 57] or increases [53, 58], whereas the number/percentage of circulating CD8+ T cytotoxic cells increases [53, 54, 58], decreases [57] or remains unchanged [56, 59]. The number/percentage of circulating B lymphocytes decreases [53] or remains unchanged following infusion of adrenaline or isoproterenol [54, 56, 59]. More recent research indicates that adrenaline increases the number of circulating CCR7−CD45RA+CD8+ effector T cells, CD4−CD8− γ/δ T cells, CD3+CD56+ NK T-like cells, CD16+CD56dim cytotoxic NK cells and CD14dimCD16+ pro-inflamma- tory monocytes. These cells most likely originate from marginated pools on the endothelial surface of blood vessels [60]. In addition to these findings, γ/δ T cells and T cells expressing chemokine receptors (CXCR2, CXCR3 and CCR5) are mobilised into the circulation following psychological stress. These responses correlate with cardiac activation [61, 62]. The effects of noradrenaline on circulating leukocytes are also variable. One study has reported that noradrenaline raised the number of circulating neutrophils, monocyte, lymphocytes and CD16+ NK cells [58, 63]. Another study found no changes in the numbers of these cell types or T lymphocyte subpopulations following treatment with noradrenaline [54]. These inconsistent findings may be due to differences between these studies in noradrenaline dose and in the duration of hormone infusion and blood sampling times relative to the period of infusion. Combined treatment with cortisol and adrenaline increases the number of circulating neutrophils for up to 12 h [52]. Growth hormone infusion in humans (2 IU) increases neutrophil number, but does not alter blood mononuclear cell subpopulations [64]. tress Hormones and Leukocyte S Function In Vivo Several of the studies described above have also examined changes in immune cell function following infusion of stress hormones in healthy humans. Cortisol does not alter Leu+ NK cell activity [51] or neutrophil chemotaxis or ­production of reactive oxygen species [65]. By contrast, adrenaline increases the activity of CD16+ NK cells [53, 55]. Similarly, noradrenaline infusion in humans (16 μg/min for 1 h) also increases CD16+ NK cell activity [63]. The effects of catecholamines and isoproterenol on lymphocyte proliferation vary. Isoproterenol reduces lymphocyte proliferation [54], whereas adrenaline and noradrenaline have no effect [54, 57]. This disparity may be due to variable changes in lymphocyte subpopulations in response to these agents. Adrenaline increases the number of T 15 Interrelations Between Acute and Chronic Exercise Stress and the Immune and Endocrine Systems cells that express IFN-γ, IL-2, IL-4 and TNF-α [53]. Adrenaline and noradrenaline infusions also raise the plasma concentrations of IL-6 and IL-1 receptor antagonist (IL-1ra) under normal resting conditions [66–68]. In contrast, adrenaline infusion prior to experimental endotoxemia reduces subsequent changes in the plasma concentrations of IL-6, IL-8 and TNF-α [69]. Hydrocortisone treatment immediately prior to experimental endotoxemia does not alter subsequent changes in plasma IL-6 concentration but attenuates plasma TNF-α concentration and increases plasma IL-10 concentration endotoxemia [70, 71]. Conversely, IL-6 and IFN-γ increase the plasma concentrations of cortisol and ACTH cortisol [72, 73], while infusion of LPS increases the plasma concentrations of adrenaline and cortisol [59]. To summarise, glucocorticoids, catecholamines and growth hormone bind to specific receptors on the surface of immune cells. This hormone-receptor binding mediates leukocyte trafficking and functional activity. In vitro, glucocorticoids and catecholamines induce a shift in the balance of type 1/type 2 cytokines towards greater production of type 2 cytokines. Growth hormone regulates immune cell activity through IGF-1 and can inhibit apoptosis of T lymphocytes. In vivo, cortisol mobilises neutrophils but reduces the number of circulating lymphocytes and does not alter circulating natural killer cell numbers. Catecholamines increase the total number of circulating lymphocytes, monocytes and natural killer cells. They also stimulate natural killer cell activity. By contrast, the effects of catecholamines on circulating lymphocyte subpopulations and lymphocyte activity are more variable. By crossing the blood–brain barrier, immune cells and cytokines can alter the function of the central nervous system. Immunoendocrine Responses to Acute Exercise Exercise immunologists have used various approaches to investigate the interaction between the endocrine and immune systems during exercise. On a basic level, some research has assessed 253 the correlation between changes in stress hormones and immunological variables following exercise. Other research has examined the interactions between the endocrine and immune systems by using different exercise workloads, carbohydrate and caffeine supplementation, thermal stress or drugs. A small number of studies have also investigated how exercise-induced immune changes alter the activity of the central nervous system. orrelations Between Stress C Hormones and Immunological Variables McCarthy et al. [74] first provided evidence that following brief, intense exercise, the number of circulating lymphocytes correlated positively with the plasma concentrations of adrenaline (ρ = 0.67, p < 0.05) and noradrenaline (ρ = 0.68, p < 0.05). Plasma adrenaline concentration also correlates positively with the number of circulating neutrophils after short, intense exercise [74, 75] and endurance exercise [76]. Rhind et al. investigated the relationships between stress hormones and immune cells following exercise. Stepwise multiple linear regression indicated that plasma adrenaline concentration accounted for some of the variation in CD3+ T cells, CD4+ T helper cells, CD8+ T cytotoxic cells and CD3−/ CD16+/CD56+ NK cells [77]. Plasma noradrenaline concentration also explained some of the variation in CD3−/CD16+/CD56+ NK cells and CD19+ B cells [77]. Steensberg et al. [78] ­discovered that following 2.5 h running at 75% VO2max (maximal oxygen uptake), the number of T helper 2 cells that produce IL-2 and IFN-γ decreases below pre-exercise values, and this response is inversely correlated with plasma adrenaline concentration. Brenner et al. [79] used stepwise multiple linear regression to examine stress hormones and immune cells following cold exposure. Plasma noradrenaline concentration accounted for some of the variation in CD3+ T cells, CD8+ T cytotoxic cells and CD19+ B cells, whereas plasma adrenaline concentration was only linked with changes in CD19+ B cells [79]. 254 The relationship between plasma cortisol concentration and the number of circulating immune cells is more variable. Some studies report no relationship [74, 80] or an inverse relationship [81] between plasma cortisol concentration and the number of circulating neutrophils after exercise. Other studies suggest that cortisol does mediate neutrophil mobilisation following exercise [76, 77, 82, 83]. The association between plasma cortisol concentration and the number of circulating monocytes following exercise is also inconsistent [77, 81]. It does seem, however, that plasma cortisol concentration accounts for some of the variation in CD4+ T helper cells and CD19+ B cells following exercise [77]. These inconsistent findings may be due to variation in blood sampling points used to examine the association between plasma cortisol concentration and the number of circulating immune cells. In contrast with adrenaline, cortisol mobilises neutrophils into the circulation in a more delayed and prolonged fashion [51, 52]. Recent evidence indicates that plasma cortisol concentration correlates strongly with lymphocyte apoptosis after resistance exercise [84]. Although growth hormone can mobilise neutrophils at rest [64], there is no clear evidence to indicate that growth hormone regulates the number of circulating neutrophils following exercise [81]. Several studies suggest that stress hormones also regulate cytokine responses to exercise. The plasma concentrations of adrenaline, noradrenaline, cortisol and growth hormone correlate with the plasma concentrations of IL-6, IL-1ra, IL-12 and TNF-α following exercise in both thermoneutral and hot conditions [85–87]. The plasma concentrations of noradrenaline and cortisol also correlate with plasma IL-6 concentration following cold exposure [79, 88]. It is unclear whether hormones or cytokines are the driving factor behind these relationships. Stress hormones and cytokines regulate body temperature during exercise, albeit through distinct mechanisms [89]. Adrenaline may stimulate a small rise in plasma IL-6 concentration during exercise [68]. Alternatively, the correlation between plasma adrenaline and IL-6 concentrations following exercise may be purely coincidental, because J. Peake both adrenaline and IL-6 regulate muscle glycogen depletion during exercise [90, 91]. IL-6 release from skeletal muscle during exercise correlates with arterial IL-6 concentration [92]. Treatment with the glucocorticoids hydrocortisone and dexamethasone reduces plasma IL-6 concentration during exercise [85]. However, IL-6 stimulates cortisol release at rest [72]. Further research is required to clarify the interactions between IL-6 and cortisol during exercise. xercise Workload, Stress Hormones E and Immunological Variables Stress hormones are released into the circulation as the intensity of exercise increases. Plasma adrenaline, noradrenaline and growth hormone concentrations rise in an exponential manner with increasing intensity [93–95]. By contrast, plasma cortisol concentration only increases above exercise intensities of >60% VO2max [76, 96, 97]. Based on these hormone responses, a number of studies have compared immunological responses to exercise of variable intensity and duration. Foster et al. [93] first provided evidence that catecholamines influence leukocyte mobilisation as a function of exercise intensity. The number of circulating granulocytes and lymphocytes increased with workload. Using the β-antagonist propranolol, they demonstrated that during exercise, catecholamines regulate changes in lymphocytes, but not granulocytes [93]. Compared with moderate-intensity exercise, the number of circulating monocytes is similar, while CD4+ T helper cells, CD8+ T cytotoxic cells and T cell proliferation decrease below pre-exercise values after high-intensity exercise [82, 97, 98]. Conversely, the number of CD19+ B cells is higher after high- vs. moderate-intensity exercise [82]. The number of circulating NK cells and NK cell activity is similar immediately after moderate- and high-intensity exercise, while NK cells and activity decrease below pre-exercise values 2 h after high-intensity exercise [98]. These studies did not evaluate the relationship between stress hormones and these intensity-dependent 15 Interrelations Between Acute and Chronic Exercise Stress and the Immune and Endocrine Systems immune changes. However, it seems likely that stress hormones play a more dominant role in mediating immune changes during high-intensity exercise. The plasma concentrations of IL-6, IL-1ra and IL-10 are also higher following highvs. moderate-intensity exercise [76, 92, 99, 100]. As discussed above, adrenaline may stimulate a minor rise in plasma IL-6 and IL-1ra concentration during exercise [66, 68], but it is more likely that IL-6 stimulates IL-1ra and IL-10 late in exercise [72]. Carbohydrate Supplementation, Stress Hormones and Immunological Variables Cortisol and adrenaline play key roles in mediating metabolism during exercise [90, 101]. Many studies have used carbohydrate supplementation to manipulate stress hormone responses and examine the mechanisms of exercise-induced changes in immune cell counts and activity. With the exception of a few studies [102–104], carbohydrate consumption during endurance exercise generally reduces the plasma concentrations of adrenaline, cortisol and growth hormone [105–112]. This decrease in the release of stress hormones most likely accounts for the decline in the number of circulating neutrophils and monocytes following carbohydrate ingestion during exercise [102, 103, 107, 109–111, 113]. By contrast, although carbohydrate supplementation attenuates plasma cortisol concentration, in general, it does not prevent the post-exercise decline in the number of circulating lymphocytes, lymphocyte subsets or NK cells [110, 114–118]. The effects of carbohydrate supplementation on other exercise-induced changes in immune cell function are variable. Despite changes in stress hormones, not all studies demonstrate that carbohydrate consumption maintains or increases neutrophil and monocyte function [102, 103, 107, 109, 113, 119]. Most research indicates that carbohydrate supplementation does not prevent the post-exercise decrease in lymphocyte proliferation [114, 118, 120]. However, Lancaster et al. [115] found that consuming carbohydrate reduces 255 plasma cortisol concentration and helps to maintain the number of IFN-γ+ CD4+ and CD8+ T cells and IFN-γ production by these cells during exercise. The metabolic stress of low muscle glycogen appears to increase plasma cortisol concentration and the number of circulating leukocytes, but does not alter lymphocyte proliferation during exercise [121, 122]. Carbohydrate supplementation increases IL-2- and IFN-γ-­ stimulated NK cell activity, but not IL-4- and IL-12-stimulated NK cell activity [116, 117]. These effects on NK cell activity are independent of changes in plasma cortisol concentration [116, 117]. Nieman et al. [123] discovered that carbohydrate ingestion during exercise reduced plasma cortisol concentration but did not alter salivary immunoglobulin A concentration (when adjusted for saliva protein concentration and secretion rate). However, changes in salivary immunoglobulin A concentration were negatively correlated with plasma cortisol concentration, and this relationship predicted the incidence of upper respiratory illness in the 2 weeks after exercise [123]. With a few exceptions [103, 106, 112], most research shows that carbohydrate attenuates the rise in plasma concentrations of IL-6, IL-10 and IL-1ra (but not IL-8 or TNF-α) following exercise [105, 108–111]. These cytokine responses to consuming carbohydrate during exercise may be partly linked to changes in catecholamine release. Carbohydrate supplementation does not influence leukocyte mRNA expression of IL-6, IL-8, IL-10 and IL-1ra or monocyte intracellular production of IL-6 and TNF-α following exercise [105, 106]. Carbohydrate ingestion attenuates the release of IL-6 from the skeletal muscle during exercise, but the effects of carbohydrate on mRNA expression of IL-6 and IL-8 in the skeletal muscle following exercise are variable [110, 111, 124, 125]. affeine Supplementation, Stress C Hormones and Immunological Variables Although caffeine is a well-known stimulant of the central nervous system, only a small number of studies have focused on its effects on stress J. Peake 256 hormones and immune responses to exercise. Ingesting 6 mg caffeine 1 h before endurance exercise consistently raises plasma adrenaline concentration [126–130]. Compared with a placebo treatment, caffeine supplementation does not alter the number of circulating neutrophils following exercise or neutrophil production of reactive oxygen species [129, 130]. The number of circulating CD3−/CD56+ NK cells is greater compared with a placebo treatment, whereas changes in the number of activated NK cells expressing CD69 are variable after exercise and caffeine ingestion [131, 132]. Changes in the total number of circulating lymphocytes after exercise and caffeine intake are also variable [129, 130]. The numbers of circulating CD4+ T helper cells and CD8+ T cytotoxic cells are lower, while the numbers of these cells that express the activation marker CD69 are greater after exercise and caffeine intake compared with a placebo treatment [126]. Caffeine supplementation also increases the concentration and secretion rate of salivary immunoglobulin A and the plasma concentration of heat shock protein 72 after exercise compared with a placebo treatment [127, 128]. This variation in the effects of caffeine on exercise-­induced immune changes may be due to differences in exercise protocol, blood sampling times and the habitual caffeine intake of the study participants. hermal Stress, Stress Hormones T and Immunological Variables Some researchers have compared changes in stress hormones and immunological variables following exercise in hot vs. cold/thermoneutral conditions. Several studies have examined responses to exercise in hot vs. cold water. This approach appears to be more effective than comparing responses to exercise in hot vs. cold/thermoneutral ambient conditions, because water is a more effective conductor of heat than air. For detailed discussion on the effects of thermal stress on the endocrine and immune systems, interested readers should consult the comprehensive review by Walsh and Whitham [89]. Plasma stress hormone concentrations are higher following exercise in hot vs. cold water, and these responses most likely account for the higher numbers of circulating neutrophils and lymphocytes following exercise in hot water [77, 81, 133–135]. However, not all research supports a link between stress hormones and the number of circulating leukocytes following exercise in hot conditions [136, 137]. This relationship may vary depending on the demands of exercise. Within the lymphocyte subsets, CD3+ T cells, CD34+ T helper cells, CD8+ T cytotoxic cells and CD3−/CD16+/CD56+ NK cells (but not CD19+ B cells) are higher at the end of exercise in hot vs. cold/thermoneutral conditions [77, 122]. By contrast, the number of circulating CD3+ T cells is lower 2 h after exercise in hot vs. thermoneutral conditions [122]. The effects of thermal stress on neutrophil function following exercise are also variable, with reports of an increase [138], a decrease [137] or no change [122, 134]. Thermal stress during exercise increases lymphocyte proliferation per cell (despite higher plasma cortisol concentration) [122], whereas it does not alter NK cell activity per cell [122, 139]. The plasma concentrations of IL-10, IL-1ra, IL-12 and TNF-α are consistently higher after exercise in hot vs. cold/thermoneutral conditions, whereas changes in the plasma concentrations of IL-6, IL-8 and granulocyte-colony-stimulating factor (G-CSF) are less consistent [86, 134, 136–138]. rugs, Stress Hormones D and Immunological Variables Several studies have used drugs to manipulate stress hormone responses to exercise and examine the resultant immunological responses. The findings of these studies are equivocal, possibly because of variation in the exercise protocols, treatment periods and drugs used in these studies. As described previously, Foster et al. [93] treated men with a single dose of the non-­ selective β1-/β2-antagonist propranolol 10 min before incremental exercise. They discovered 15 Interrelations Between Acute and Chronic Exercise Stress and the Immune and Endocrine Systems that during exercise, propranolol reduced the rise in the number of circulating lymphocytes, but not neutrophils or plasma catecholamine concentrations. This finding suggests that catecholamines may not regulate leukocyte mobilisation directly during incremental exercise. Instead, catecholamines may work indirectly by increasing blood flow, which strips leukocytes from the endothelial surface of blood vessels in marginal pools such as the lungs. Murray et al. [140] conducted a follow-up study in which they treated men and women with propranolol or the selective β1-­ antagonist metoprolol for 1 week prior to an incremental exercise test. Neither drug reduced post-exercise plasma catecholamine concentrations compared with the control trial. However, compared with the control trial, propranolol (but not metoprolol) reduced the total number of circulating lymphocytes, numbers of CD4+ T helper cells and CD8+ T cytotoxic cells and NK cell numbers and activity and reduced the post-­ exercise decline in lymphocyte proliferation [140]. These findings suggest that circulating catecholamines may not mobilise lymphocytes into the circulation. Instead, these cells may be mobilised from the spleen in response to direct activation of β1-/β2-adrenergic receptors in the spleen [141]. Starkie et al. [142] treated men with the selective α1-antagonist prazosin and the non-selective β-antagonist timolol or placebo 2 h prior to 20 min cycling at ∼78% VO2max. Plasma catecholamine concentrations were higher, whereas plasma cortisol concentration was lower after exercise in the drug trial compared with the placebo trial. Starkie et al. [142] attributed the greater catecholamine response in the drug trial to reduced clearance of catecholamines by β-receptors. The numbers of circulating lymphocytes and monocytes increased during exercise in both trials but were lower immediately after exercise in the drug trial compared with the placebo trial—despite the higher plasma catecholamine concentrations. This finding conflicts with other research showing that infusion of adrenaline or isoproterenol raises the number of circulating lymphocytes [51, 53, 54]. One possible explanation for this difference is that the drugs used in 257 the study by Starkie et al. [142] may target different adrenergic receptors on lymphocyte compared with adrenaline or isoproterenol. The numbers of circulating IFN-γ+ CD3+ T cells, IL-2+ CD3+ T cells and IFN-γ+ CD3−/CD56+ NK cells increased during exercise in both trials. However, the numbers of these cells were lower after exercise in the drug trial compared with the placebo trial. IL-2 production by CD3+ T cells and IFN-γ production by both IFN-γ+ CD3+ T cells and IFN-γ+ CD3−/CD56+ NK cells decreased during exercise similarly in both trials. These findings suggest that α- and/or β-adrenergic receptor stimulation does not regulate cytokine production by T cells and NK cells during exercise. Mazzeo et al. [143] treated women with prazosin or placebo for 3 days before cycling for 50 min at 50% VO2max. Prazosin reduced plasma IL-6 concentration after exercise compared with the placebo. Papanicolaou et al. [85] treated men with hydrocortisone, dexamethasone or a placebo 4 h before 25 min running at 78% VO2max. Both hydrocortisone and dexamethasone attenuated plasma IL-6 concentration after exercise compared with the placebo. vidence for Interactions Between E the Central Nervous and Immune Systems As outlined above, considerable attention has focused on how stress hormones regulate immune responses to exercise. The immune system is also capable of altering the function of the central nervous system. Several studies have examined this issue, and it is likely that more research will be conducted in this area in the future. In mice, exercise-­induced muscle damage stimulates macrophages residing in the brain to secrete IL-1β into the surrounding tissue [144, 145]. This response appears to increase perceptions of fatigue, reduce voluntary activity and delay recovery from exercise [146]. In humans, Steensberg et al. [147] observed that at rest, the concentrations of IL-6 and the cellular chaperone heat shock protein 72 (HSP72) are two to three 258 times higher in cerebrospinal fluid compared with plasma. Although exercise stimulates the systemic release of IL-6 and heat shock protein 72, their concentrations remain stable in cerebral spinal fluid, which indicates that they do not cross the blood–brain barrier [147]. The brain releases small amounts of IL-6 into the systemic circulation during exercise, and this is independent of hyperthermia [148]. The functional significance of this response is not certain. It may provide a signal to the liver to increase glucose output, or it may be a more general indication of increased neural activity during exercise [148]. hronic Interactions Between C the Endocrine and Immune Systems Compared with the amount of research on acute exercise, fewer studies have examined interactions between stress hormones and immunological variables following chronic training. Most studies have simply documented the effects of intensified training on simultaneous changes in stress hormones and immune cell counts at rest and/or in response to acute exercise. Very few studies have specifically examined the relationship between changes in stress hormones and immune cell counts and function. Several studies report no changes in resting plasma and urinary cortisol concentrations, immune cell counts or serum cytokine concentrations after intensified training [149–153]. Robson-Ansley et al. [154] discovered no changes in resting plasma cortisol or the number of circulating neutrophil counts but did find that resting plasma IL-6 concentration was persistently elevated following 4 weeks of intense training. Fry et al. [155] observed that resting plasma cortisol concentration decreased, while the numbers of circulating neutrophils, monocytes and lymphocytes did not change after 10 days of intense interval training. The number of circulating CD3+, CD4+ and CD8+ T cells and CD20+ B cells also remained unchanged, whereas the number of circulating CD56+ NK cells decreased and CD25+ T cells increased following 10 days of training [155]. It is unlikely, however, J. Peake that these changes in CD56+ NK cells and CD25+ T cells were related to changes in plasma cortisol concentration. Smith and Myburgh [156] reported no change in resting plasma cortisol concentration but found that CD4+ and CD8+ T cell counts and CD16+/CD56+ NK cells decreased following 4 weeks of intense training. Makras et al. [157] observed an increase in urinary cortisol concentration, an increase in CD4+ T cell count and a decrease in neutrophil count at rest after 4 weeks of military training. Ortega et al. [158] found that neutrophil phagocytic activity was higher in female athletes compared with non-athletes. In the athletes, neutrophil phagocytic activity correlated positively with plasma cortisol concentration, whereas it correlated negatively with plasma ACTH concentration. Findings from the study by Cunniffe et al. [159] suggest that elevated salivary cortisol concentration with training may reduce salivary immunoglobulin A concentration, resulting in increased susceptibility to upper respiratory illness. Some of the variability among these studies may result from differences in the physical fitness of study participants, training loads and blood sampling times. The effects of chronic training on cortisol and immune responses to acute exercise are also ­variable. Verde et al. [160] reported that changes in serum cortisol concentration, CD3+ T cell counts and lymphocyte proliferation after acute exercise were all attenuated following 3 weeks of intense training. Lancaster et al. [161] discovered that 2 weeks of intense training reduced plasma cortisol concentration but did not alter lymphocyte production of the type 1 cytokine IFN-γ or the type 2 cytokine IL-4. In contrast with these findings, other research indicates no effect of chronic training on exercise-induced changes in plasma and salivary cortisol concentration, immune cell counts or salivary immunoglobulin A concentration [150, 152, 162]. A small number of studies have examined changes in plasma or urinary catecholamine concentrations and immune cell counts following chronic training. Imrich et al. [150] found no changes in plasma catecholamine concentration or immune cell counts at rest or in response to acute exercise following 6 weeks of training. 15 Interrelations Between Acute and Chronic Exercise Stress and the Immune and Endocrine Systems Hooper et al. [163] reported that both the number of circulating neutrophils and plasma noradrenaline concentration were elevated in swimmers showing symptoms of overtraining compared with swimmers who were not overtrained after 6 months of training. However, it is unclear whether these responses were linked in any way. Mackinnon et al. [151] observed that urinary norepinephrine concentration decreased, whereas plasma noradrenaline and leukocyte counts at rest did not change following 4 weeks of intense training. Makras et al. [157] found that the ratio of adrenaline to noradrenaline in urine increased after 4 weeks of military training. This response correlated positively with CD4+ T cell counts and correlated negatively with neutrophil counts. Biological Significance of Interactions Between the Endocrine and Immune Systems Dhabhar [1] proposes the following analogy to explain the possible significance of acute stress on the immune system. Within minutes of the onset of stress, catecholamines stimulate the body’s ‘soldiers’ (i.e., leukocytes) to leave their ‘barracks’ (i.e., spleen, lung, bone marrow, lymph nodes) and enter the ‘boulevards’ (i.e., blood vessels and lymphatics). As stress proceeds, glucocorticoids are released which stimulate leukocytes to exit the bloodstream and enter potential ‘battle stations’ (i.e., skin, lung, gastrointestinal and urinary-­ genital tracts, mucosal surfaces and lymph nodes) in preparation for immune challenges that may occur in response to stressful stimuli [1]. In the context of exercise, the factors that stimulate the release of stress hormones are most often non-harmful. These factors may include demands for (1) increased blood flow to contracting muscle (to deliver oxygen and nutrients) and skin (for thermoregulation) and (2) release of energy substrates from the liver and adipose tissue (e.g., glucose, fatty acids, amino acids) to support muscle metabolism. Interaction between the endocrine and immune systems during exercise can therefore be considered as rather non-­ 259 specific. However, stress hormones may (incidentally) prime immune cells to respond to infectious pathogens and/or airborne pollutants that invade mucosal surfaces lining the respiratory tract. Dhabhar [1] proposed that the effects of stress on the immune system and general health depend on the duration of exposure to stress. Acute and intense stress may enhance immune function, and mild stress of moderate duration may promote immunosurveillance, while chronic stress may cause immune dysregulation [1]. Immunoprotection resulting from acute stress may lead to more effective wound healing, responses to vaccination and resistance to infection and cancer. Immunopathology resulting from severe acute stress or persistent stress may promote pro-inflammatory and autoimmune diseases. Immunosuppression resulting from chronic stress may reduce the effectiveness of wound healing and vaccination and increase the risk of infection and cancer. By contrast, chronic stress may reduce the risk of pro-inflammatory and autoimmune diseases by suppressing aspects of immune function that contribute to such conditions (e.g., T lymphocyte activity, cytokine production) [1]. Both acute exercise [164] and chronic training [165, 166] increase antibody production in response to vaccination. Mild repeated stress resulting from chronic training also improves the rate of wound healing [167], decreases the risk of upper respiratory illness [168] and reduces the prevalence and severity of various chronic diseases [169]. Although more work is needed to define their precise role, it is likely that stress hormones mediate some of these benefits of exercise. Summary A variety of non-harmful stimuli during exercise induce the release of stress hormones. These stress hormones influence many physiological systems, including the immune system. Stress hormones act to mobilise immune cells into the circulation and can increase or decrease the activity of these cells. The precise nature of the inter- 260 action between stress hormones and immune cells likely depends on multiple factors, including the intensity and duration of exercise, the physical fitness of exercising individuals and environmental conditions. Some stress hormones (e.g., catecholamines) influence immune cell activity mainly during exercise, whereas others (e.g., cortisol) may have a more delayed effect on immune function during the later stages of exercise and/or after exercise. Nutritional interventions such as carbohydrate and caffeine supplementation can alter the secretion of stress hormones during exercise, but these alterations do not always result in changes in immune function. Immunoendocrine interactions during exercise may serve to promote some aspects of health. However, further research is needed to understand the biological significance of such interactions in more detail. References 1. Dhabhar FS. Enhancing versus suppressive effects of stress on immune function: implications for immunoprotection and immunopathology. Neuroimmunomodulation. 2009;16:300–17. 2. Glaser R, Kiecolt-Glaser JK. Stress-induced immune dysfunction: implications for health. Nat Rev Immunol. 2005;5:243–51. 3. Bartholome B, Spies CM, Gaber T, Schuchmann S, Berki T, Kunkel D, et al. Membrane glucocorticoid receptors (mGCR) are expressed in normal human peripheral blood mononuclear cells and up-regulated after in vitro stimulation and in patients with rheumatoid arthritis. FASEB J. 2004;18:70–80. 4. Miller AH, Spencer RL, Pearce BD, Pisell TL, Azrieli Y, Tanapat P, et al. Glucocorticoid receptors are differentially expressed in the cells and tissues of the immune system. Cell Immunol. 1998;186:45–54. 5. Maisel AS, Harris T, Rearden CA, Michel MC. Beta-­ adrenergic receptors in lymphocyte subsets after exercise. Alterations in normal individuals and patients with congestive heart failure. Circulation. 1990;82:2003–10. 6. Liggett SB. Identification and characterization of a homogeneous population of beta 2-adrenergic receptors on human alveolar macrophages. Am Rev Respir Dis. 1989;139:552–5. 7. Marinetti GV, Rosenfeld SI, Thiem PA, Condemi JJ, Leddy JP. Beta-adrenergic receptors of human leukocytes. Studies with intact mononuclear and polymorphonuclear cells and membranes comparing two radioligands in the presence and absence of chloroquine. Biochem Pharmacol. 1983;32:2033–43. J. Peake 8. Swanson MA, Lee WT, Sanders VM. IFN-gamma production by Th1 cells generated from naive CD4+ T cells exposed to norepinephrine. J Immunol. 2001;166:232–40. 9. Sanders VM, Baker RA, Ramer-Quinn DS, Kasprowicz DJ, Fuchs BA, Street NE. Differential expression of the beta2-adrenergic receptor by Th1 and Th2 clones: implications for cytokine production and B cell help. J Immunol. 1997;158:4200–10. 10. Loza MJ, Foster S, Peters SP, Penn RB. Beta-agonists modulate T-cell functions via direct actions on type 1 and type 2 cells. Blood. 2006;107:2052–60. 11. Bao JY, Huang Y, Wang F, Peng YP, Qiu YH. Expression of alpha-AR subtypes in T lymphocytes and role of the alpha-ARs in mediating modulation of T cell function. Neuroimmunomodulation. 2007;14:344–53. 12. Roupe van der Voort C, Heijnen CJ, Wulffraat N, Kuis W, Kavelaars A. Stress induces increases in IL-6 production by leucocytes of patients with the chronic inflammatory disease juvenile rheumatoid arthritis: a putative role for alpha(1)-adrenergic receptors. J Neuroimmunol. 2